A Dissertation entitled

Marinobufagenin Induced Uremic Cardiomyopathy: The Role of Passive Immunization, Rapamycin, and CD40 Signaling in The Generation of Renal Fibrosis

By

Steven T. Haller

Submitted to the Graduate Faculty as partial fulfillment of the requirements for the

Doctor of Philosophy Degree in Biomedical Sciences

Dr. Joseph I. Shapiro, Committee Chair

Dr. Christopher J. Cooper, Committee Member

Dr. Deepak Malhotra, Committee Member

Dr. Zijian Xie, Committee Member

Dr. Jiang Liu, Committee member

Dr. Patricia R. Komuniecki, Dean College of Graduate Studies

The University of Toledo August 2012

Copyright 2012, Steven Thomas Haller This document is copyrighted material. Under copyright law, no parts of this document may be reproduced without the expressed permission of the author. An Abstract of Marinobufagenin Induced Uremic Cardiomyopathy: The Role of Passive Immunization, Rapamycin, and CD40 Signaling in The Generation of Renal Fibrosis

By Steven T. Haller Submitted to the Graduate Faculty as partial fulfillment of the requirements for the

Doctor of Philosophy Degree in Biomedical Sciences

The University of Toledo

August 2012

Cardiotonic such as marinobufagenin (MBG) and are specific ligands for the Na/K-ATPase and represent a relatively new class of hormones.

Uremic cardiomyopathy is characterized by a decrease in diastolic function, left ventricular hypertrophy, oxidant stress, and both cardiac and renal fibrosis. We have shown that MBG, signaling through the Na/K-ATPase, causes many of the adverse pathological effects of experimental uremic cardiomyopathy induced by 5/6th

nephrectomy (PNx) in the rat. The goal of this dissertation is to describe some of the

manipulations we have performed in order to provide potential therapies for the treatment

iii

of uremic cariomyopathy. Specifically, we show that treatment with an anti-MBG antibody drastically reduces cardiac fibrosis in PNx animals. Treatment with rapamycin

(an mTOR inhibitor) produced similar effects with the added benefit of reducing circulating MBG in these animals. In addition, we show that ischemic renal disease is accompanied with elevated levels of the platelet activation marker soluble CD40 ligand

(sCD40L), and its soluble receptor, CD40, may predict outcomes in this disease state.

Data in our PNx model suggests a role for proximal tubular CD40 activation contributing to the development of renal fibrosis, which may be potentiated by cardiotonic steroid signaling through the Na/K-ATPase.

iv

Dedication

To my family who has faithfully stood by my side throughout my academic career. Your undying love and support has inspired me to be a better person. I am forever grateful for the sacrifices you have made for me along the way. Thank you for believing in me. I love you all.

v

Acknowledgments

I am extremely grateful to Drs. Joseph I. Shapiro and Christopher J. Cooper for their

tremendous guidance and faithful support throughout my graduate studies. You have

inspired my passion for translational research. I look forward to working with you in the

future.

I would like to thank my academic advisory committee members: Dr. Zijian Xie, Dr.

Deepak Malhotra, and Dr. Jiang Liu, for there expert advice and guidance throughout my

graduate studies.

Dr. Liu, Thank you for letting me waste away hours in your office discussing my work. I look forward to working with you in the future.

To Dr. Cooper and Holly Burtch for teaching me that being involved in human subjects

research is a privilege, not a right.

To the Clinical Coordinating Center staff, Thank you for supporting and believing in me.

To Pam Brewster, Thank you for your expert advice and support with statistical analysis.

To Carol Woods, Thank you for all of your support.

To Dr. Periyasamy who taught me to walk before I run. I miss you every day.

To Dr. Kennedy, My mentor, my friend, my other brother.

vi

Table of Contents

Abstract iii

Dedication V

Acknowledgments Vi

Table of Contents Vii

Chapter 1 – Literature Review and Introduction 1

Chapter 2 – “Monoclonal antibody against marinobufagenin reverses cardiac fibrosis in

rats with chronic renal failure” (Manuscript) 10

2.1 Abstract 11

2.2 Introduction 12

2.3 Methods 14

2.4 Results 19

2.5 Discussion 20

2.6 Manuscript References 23

2.7 Table and Figure Legends 27 vii

2.8 Table and Figures 29

Chapter 3 – “Rapamycin reduces cardiac fibrosis in experimental uremic cardiomyopathy” (Manusript to be submitted) 33

3.1 Abstract 34

3.2 Introduction 36

3.3 Methods 37

3.4 Results 42

3.5 Discussion 44

3.6 Manuscript References 47

3.7 Table and Figure Legends 52

3.8 Table and Figures 54

Chapter 4 – “Platelet activation in patients with atherosclerotic renal artery stenosis undergoing stent revascularization” (Manuscript) 59

4.1 Abstract 60

4.2 Introduction 61

4.3 Materials and Methods 62

4.4 Results 64

4.5 Discussion 66 viii

4.6 Manuscript References 71

4.7 Table and Figure Legends 78

4.8 Table and Figures 79

Chapter 5 – CD40 mediated fibrosis in chronic and ischemic renal disease 83

5.1 Chronic Kidney Disease and Renal Ischemia 83

5.2 Platelet Activation, CD40 Signaling, and Fibrosis 85

5.3 Preliminary Data: Clinical Trial 86

5.4 Preliminary Data: Animal Studies 90

5.5 Preliminary Data: LLC-PK1 Cells 94

5.6 Conclusions 99

5.7 Chapter 5 References 101

Chapter 6 – Summary and Conclusions 110

ix

Chapter 1-Literature Review and Introduction

1.1 Structure and Function of the Na/K-ATPase

The Na/K-ATPase, originally discovered by Skou (1957), is a member of the P-

type ATPase family, and plays an essential role in regulating the cellular transmembrane

ion gradient by ATP-dependent transport of Na+ and K+ across the plasma membrane.1, 2

In order to maintain ion transport the Na/K-ATPase is in constant flux between two major conformation states, E1 and E2.3 The E1 state has high affinity for Na+ and ATP, while

the E2 state has high affinity for K+.3 The Na/K-ATPase is composed of two

noncovalently linked subunits, α and β which together form the functional unit of the

enzyme.2 There are four α subunit isoforms (α1, α2, α3, and α4), and three β subunit

isoform (β1, β2, and β3) all expressed in a tissue specific mannor.4 The α subunit is the catalytic subunit, which contains specific binding sites for Na+, K+, ATP, and cardiotonic steroids (CTS, ligands of the Na/K-ATPase). The β subunit plays an essential role in regulating the activity of the enzyme.2 A third subunit (γ) contains the conserved FXYD

motif.3 The γ subunit is not considered to be essential for enzymatic function, but has

been proposed to modulate enzymatic activity.5, 6

In addition to the essential ion pumping function, elegant work from the

laboratory of Dr. Xie and collaborators has shown that the cardiotonic steroid ouabain

binds to the α1 subunit of the Na/K-ATPase converting it into a signal transducer capable of activating multiple protein kinase cascades.7-9 Src binds to the Na/K-ATPase α1 1

subunit forming a functional signaling complex 10. CTS bind to the Na/K-ATPase and induce a conformational change which activates Src 10. Src transactivates the epidermal

growth factor receptor (EGFR) which results in the activation of phospholipase C (PLC),

phosphoinsitide 3-kinase (PI3K), mitogen-activated protein kinases (MAPKs), protein kinase C (PKC), and the generation of reactive oxygen species (ROS). 4, 9 Importantly,

ouabain binding to the Na/K-ATPase induces endocytosis of the receptor complex in a

manner consistent with classic receptor tyrosine kinases.11

1.2 Cardiotonic Steroids and Uremic Cardiomyopathy

CTS are specific ligands for the Na/K-ATPase, and have been used to treat heart failure by coupling reduced Na/K-ATPase activity with a reduction in Na+/ Ca2+-

exchanger activity ultimately leading to an accumulation of intracellular sodium and

increases in cytosolic calcium.3, 4 This increase in cytosolic calcium results in increased

cardiac contractile function.4 Endogenous CTS, such as ouabain and marinobufagenin

(MBG), represent a relatively new class of steroid hormones. Endogenous ouabain has

been postulated to be produced from the adrenal cortex and hypothalamus.4 In

, the biosynthesis of MBG has been proposed to occur via the bile acid

pathway from cholic acids.4 Elevated levels of endogenous CTS have been reported in a

variety of clinical conditions associated with plasma volume expansion such as

congestive heart failure, chronic renal failure, hypertension, renal ischemia, and

preeclamsia.12-19 In animal models, administration of ouabain and MBG have been

shown to cause hypertension, cardiac hypertrophy, and fibrosis.20-22 Furthermore, salt-

loading in Dahl salt-sensitive rats caused an increase in brain derived ouabain, which

2

elevated plasma MBG levels contributing to hypertension in a process mediated by

angiotension II.23

Recent data indicates that chronic kidney disease (CKD) is prevalent, affecting up

to 11% of the US adult population. 24 Platelet activation and inflammation have been

implicated in the progression CKD. 25 Cardiovascular disease is both common and a

major cause of mortality in patients with CKD. 26, 27 This uremic cardiomyopathy is

characterized by a decrease in diastolic function, left ventricular hypertrophy, oxidant

stress, and both cardiac and renal fibrosis. 4, 22, 28 We have shown that MBG, signaling

through the Na/K-ATPase, causes many of the adverse pathological effects of

experimental uremic cardiomyopathy induced by 5/6th nephrectomy (PNx) in the rat. 21

Our group has demonstrated that pharmacologic administration of MBG causes cardiac hypertrophy and fibrosis, as seen in patients, whereas active immunization against MBG attenuated this in PNx. 21, 22 Additionally, cardiac fibroblasts treated with MBG, at

concentrations similar to those reported in experimental and clinical renal failure, has

been shown to stimulate collagen production. 22 This increase in collagen production

appears to be dependent on the Na/K-ATPase-Src-EGFR-ROS signaling cascade. 22 The

transcription factor Friend leukemia integration-1 (Fli-1) has been shown to be a negative regulator of collagen synthesis. 29, 30 PKC- δ phosphorylates Fli-1 and promotes collagen

synthesis. 31 We have shown that MBG signaling through the Na/K-ATPase, caused

PKC- δ translocation to the nucleus leading to Fli-1 phosphorylation and collagen

production. 32 A recent report from our lab has shown that treatment with a monoclonal

antibody directed against MBG (3E9 mAb) in PNx animals resulted in a drastic decrease

in blood pressure, significantly reduced cardiac levels of oxidant stress, increased the 3

expression of Fli-1, and caused a significant reduction in cardiac fibrosis.33

Spironolactone and its major metabolite have been shown to competitively inhibit CTS binding to the Na/K-ATPase.34 We have shown that spironolactone treatment in both

PNx animals and animals receiving MBG infusion attenuated diastolic dysfunction and cardiac fibrosis in these experimental animal models.35 Our findings indicate that spironolactone and the 3E9 monoclonal antibody may offer potential indications for the treatment of uremic cardiomyopathy.

4

References for Literature Review and Introduction

1. Skou JC. The influence of some cations on an adenosine triphosphatase from

peripheral nerves. Biochim Biophys Acta. 1957;23(2):394-401.

2. Kaplan JH. Biochemistry of Na,K-ATPase. Annu Rev Biochem. 2002;71:511-535.

3. Yatime L, Laursen M, Morth JP, Esmann M, Nissen P, Fedosova NU. Structural

insights into the high affinity binding of cardiotonic steroids to the Na+,K+-

ATPase. J Struct Biol.174(2):296-306.

4. Bagrov AY, Shapiro JI, Fedorova OV. Endogenous cardiotonic steroids:

physiology, pharmacology, and novel therapeutic targets. Pharmacol Rev.

2009;61(1):9-38.

5. Arystarkhova E, Wetzel RK, Asinovski NK, Sweadner KJ. The gamma subunit

modulates Na(+) and K(+) affinity of the renal Na,K-ATPase. J Biol Chem.

1999;274(47):33183-33185.

6. Geering K. Function of FXYD proteins, regulators of Na, K-ATPase. J Bioenerg

Biomembr. 2005;37(6):387-392.

7. Xie Z, Askari A. Na(+)/K(+)-ATPase as a signal transducer. Eur J Biochem.

2002;269(10):2434-2439.

8. Xie Z, Cai T. Na+-K+--ATPase-mediated signal transduction: from protein

interaction to cellular function. Mol Interv. 2003;3(3):157-168.

9. Xie Z. Molecular mechanisms of Na/K-ATPase-mediated signal transduction.

Ann N Y Acad Sci. 2003;986:497-503.

5

10. Tian J, Cai T, Yuan Z, Wang H, Liu L, Haas M, Maksimova E, Huang XY, Xie

ZJ. Binding of Src to Na+/K+-ATPase forms a functional signaling complex. Mol

Biol Cell. 2006;17(1):317-326.

11. Liu J, Kesiry R, Periyasamy SM, Malhotra D, Xie Z, Shapiro JI. Ouabain induces

endocytosis of plasmalemmal Na/K-ATPase in LLC-PK1 cells by a clathrin-

dependent mechanism. Kidney Int. 2004;66(1):227-241.

12. Komiyama Y, Dong XH, Nishimura N, Masaki H, Yoshika M, Masuda M,

Takahashi H. A novel endogenous digitalis, telocinobufagin, exhibits elevated

plasma levels in patients with terminal renal failure. Clin Biochem.

2005;38(1):36-45.

13. Rossi G, Manunta P, Hamlyn JM, Pavan E, De Toni R, Semplicini A, Pessina

AC. Immunoreactive endogenous ouabain in primary aldosteronism and essential

hypertension: relationship with plasma renin, aldosterone and blood pressure

levels. J Hypertens. 1995;13(10):1181-1191.

14. Gonick HC, Ding Y, Vaziri ND, Bagrov AY, Fedorova OV. Simultaneous

measurement of marinobufagenin, ouabain, and hypertension-associated protein

in various disease states. Clin Exp Hypertens. 1998;20(5-6):617-627.

15. Fedorova OV, Talan MI, Agalakova NI, Lakatta EG, Bagrov AY. Endogenous

ligand of alpha(1) sodium pump, marinobufagenin, is a novel mediator of sodium

chloride--dependent hypertension. Circulation. 2002;105(9):1122-1127.

16. Fridman AI, Matveev SA, Agalakova NI, Fedorova OV, Lakatta EG, Bagrov AY.

Marinobufagenin, an endogenous ligand of alpha-1 sodium pump, is a marker of

congestive heart failure severity. J Hypertens. 2002;20(6):1189-1194.

6

17. Tian J, Haller S, Periyasamy S, Brewster P, Zhang H, Adlakha S, Fedorova OV,

Xie ZJ, Bagrov AY, Shapiro JI, Cooper CJ. Renal ischemia regulates

marinobufagenin release in humans. Hypertension.56(5):914-919.

18. Lopatin DA, Ailamazian EK, Dmitrieva RI, Shpen VM, Fedorova OV, Doris PA,

Bagrov AY. Circulating bufodienolide and sodium pump inhibitors in

preeclampsia. J Hypertens. 1999;17(8):1179-1187.

19. Kolmakova EV, Haller ST, Kennedy DJ, Isachkina AN, Budny GV, Frolova EV,

Piecha G, Nikitina ER, Malhotra D, Fedorova OV, Shapiro JI, Bagrov AY.

Endogenous cardiotonic steroids in chronic renal failure. Nephrol Dial

Transplant.26(9):2912-2919.

20. Kimura K, Manunta P, Hamilton BP, Hamlyn JM. Different effects of in vivo

ouabain and on renal artery function and blood pressure in the rat.

Hypertens Res. 2000;23 Suppl:S67-76.

21. Kennedy DJ, Vetteth S, Periyasamy SM, Kanj M, Fedorova L, Khouri S, Kahaleh

MB, Xie Z, Malhotra D, Kolodkin NI, Lakatta EG, Fedorova OV, Bagrov AY,

Shapiro JI. Central role for the cardiotonic steroid marinobufagenin in the

pathogenesis of experimental uremic cardiomyopathy. Hypertension.

2006;47(3):488-495.

22. Elkareh J, Kennedy DJ, Yashaswi B, Vetteth S, Shidyak A, Kim EG, Smaili S,

Periyasamy SM, Hariri IM, Fedorova L, Liu J, Wu L, Kahaleh MB, Xie Z,

Malhotra D, Fedorova OV, Kashkin VA, Bagrov AY, Shapiro JI.

Marinobufagenin stimulates fibroblast collagen production and causes fibrosis in

experimental uremic cardiomyopathy. Hypertension. 2007;49(1):215-224.

7

23. Fedorova OV, Agalakova NI, Talan MI, Lakatta EG, Bagrov AY. Brain ouabain

stimulates peripheral marinobufagenin via angiotensin II signalling in NaCl-

loaded Dahl-S rats. J Hypertens. 2005;23(8):1515-1523.

24. Kronenberg F. Emerging risk factors and markers of chronic kidney disease

progression. Nat Rev Nephrol. 2009;5(12):677-689.

25. Tu X, Chen X, Xie Y, Shi S, Wang J, Chen Y, Li J. Anti-inflammatory

renoprotective effect of clopidogrel and irbesartan in chronic renal injury. J Am

Soc Nephrol. 2008;19(1):77-83.

26. Chade AR, Lerman A, Lerman LO. Kidney in early atherosclerosis.

Hypertension. 2005;45(6):1042-1049.

27. Jaradat MI, Molitoris BA. Cardiovascular disease in patients with chronic kidney

disease. Semin Nephrol. 2002;22(6):459-473.

28. Fedorova LV, Raju V, El-Okdi N, Shidyak A, Kennedy DJ, Vetteth S,

Giovannucci DR, Bagrov AY, Fedorova OV, Shapiro JI, Malhotra D. The

cardiotonic steroid hormone marinobufagenin induces renal fibrosis: implication

of epithelial-to-mesenchymal transition. Am J Physiol Renal Physiol.

2009;296(4):F922-934.

29. Czuwara-Ladykowska J, Shirasaki F, Jackers P, Watson DK, Trojanowska M. Fli-

1 inhibits collagen type I production in dermal fibroblasts via an Sp1-dependent

pathway. J Biol Chem. 2001;276(24):20839-20848.

30. Wang Y, Fan PS, Kahaleh B. Association between enhanced type I collagen

expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts.

Arthritis Rheum. 2006;54(7):2271-2279.

8

31. Jinnin M, Ihn H, Yamane K, Mimura Y, Asano Y, Tamaki K. Alpha2(I) collagen

gene regulation by protein kinase C signaling in human dermal fibroblasts.

Nucleic Acids Res. 2005;33(4):1337-1351.

32. Elkareh J, Periyasamy SM, Shidyak A, Vetteth S, Schroeder J, Raju V, Hariri IM,

El-Okdi N, Gupta S, Fedorova L, Liu J, Fedorova OV, Kahaleh MB, Xie Z,

Malhotra D, Watson DK, Bagrov AY, Shapiro JI. Marinobufagenin induces

increases in procollagen expression in a process involving protein kinase C and

Fli-1: implications for uremic cardiomyopathy. Am J Physiol Renal Physiol.

2009;296(5):F1219-1226.

33. Haller ST, Kennedy DJ, Shidyak A, Budny GV, Malhotra D, Fedorova OV,

Shapiro JI, Bagrov AY. Monoclonal Antibody Against Marinobufagenin Reverses

Cardiac Fibrosis in Rats With Chronic Renal Failure. Am J Hypertens. March

2012 [epub ahead of print].

34. Finotti P, Palatini P. Canrenone as a partial agonist at the digitalis receptor site of

sodium-potassium-activated adenosine triphosphatase. J Pharmacol Exp Ther.

1981;217(3):784-790.

35. Tian J, Shidyak A, Periyasamy SM, Haller S, Taleb M, El-Okdi N, Elkareh J,

Gupta S, Gohara S, Fedorova OV, Cooper CJ, Xie Z, Malhotra D, Bagrov AY,

Shapiro JI. Spironolactone attenuates experimental uremic cardiomyopathy by

antagonizing marinobufagenin. Hypertension. 2009;54(6):1313-1320.

9

Chapter 2 – Manuscript

Title: Monoclonal Antibody Against Marinobufagenin Reverses Cardiac Fibrosis in Rats With Chronic Renal Failure

Authors:

Steven T. Haller1, David J. Kennedy1,2, Amjad Shidyak2, George V. Budny1, Deepak Malhotra1,Olga V. Fedorova3, Joseph I. Shapiro1 and Alexei Y. Bagrov3

1College of Medicine, University of Toledo, Toledo, Ohio, USA;

2Departmentof Cell Biology, Cleveland Clinic Lerner Research Institute, Cleveland,

Ohio,USA; 3Laboratory of Cardiovascular Science, National Institute on Aging,

NIH,Baltimore, Maryland, USA.

Corresponding Author:

Dr. Alexei Y. Bagrov at the National Institute on Aging, NIH, 5600 Nathan Shock Drive,

Baltimore, MD 21224

[email protected]

Published in American Journal of Hypertension. March 2012 1. doi:10.1038/ajh.2012.17.

[Epub ahead of print]

10

2.1 Abstract

Cardiotonic steroids (CTS) are implicated in pathophysiology of uremic cardiomyopathy. In the present study, we tested whether a monoclonal antibody (mAb) against the CTS, marinobufagenin (MBG), alleviates cardiac hypertrophy and fibrosis in partially nephrectomized (PNx) rats. In PNx rats, we compared the effects of 3E9 anti-MBG mAb and of Digibind, an affinity-purified digoxin antibody, on blood pressure and cardiac hypertrophy and fibrosis following 4 weeks after the surgery. In

PNx rats, a fourfold elevation in plasma MBG levels was associated with hypertension, increased cardiac levels of carbonylated protein, cardiac hypertrophy, a reduction in cardiac expression of a nuclear transcription factor which is a negative regulator of collagen synthesis, Friend leukemia integration-1 (Fli-1), and an increase in the levels of collagen-1. A single intraperitoneal administration of 3E9 mAb to PNx rats reduced blood pressure by 59 mm Hg for 7 days and produced a significant reduction in cardiac weight and cardiac levels of oxidative stress, an increase in the expression of Fli-1, and a reduction in cardiac fibrosis. The effects of Digibind were similar to those of 3E9 mAb, but were less pronounced. In experimental chronic renal failure, elevated levels of MBG contribute to hypertension and induce cardiac fibrosis via suppression of Fli-1, representing a potential target for therapy.

11 2.2 Introduction

Uremic cardiomyopathy is a major cause of morbidity and mortality in patients

with chronic kidney disease.1 Despite considerable recent progress in the understanding

of the pathogenesis of uremic cardiomyopathy, there is clearly a niche for novel

approaches to its treatment.1,2 An increasing body of evidence indicates that one of the

factors implicated in pathogenesis of uremic cardiomyopathy is the group of hormones

known as endogenous cardiotonic steroids (CTS).3 CTS regulate sodium pump activity

at a cellular level and are implicated in the regulation of natriuresis and vascular tone.3

Many of the effects of these hormones appear to derive from a signaling function of the

Na/K-ATPase; in particular, this signaling stimulated by CTS leads to cardiac

hypertrophy and fibrosis.4,5

Previously, we demonstrated that circulating concentrations of marinobufagenin

(MBG) (14,15β-Epoxy-3β,5-dihydroxy-5β-bufa-20,22 dienolide), an endogenous bufadienolide CTS, are elevated in patients with renal failure and in partially nephrectomized rats (PNx).5,6 In PNx rats, also we observed increased cardiac and

plasma levels of carbonylated proteins as well as other evidence for signaling through the

Na/K-ATPase such as activation of Src and mitogen-activated protein kinase (MAPK).5,6

In these studies, active immunization of PNx rats against MBG dramatically reduced cardiac hypertrophy and fibrosis and systemic oxidant stress, as well as evidence of

Na/K-ATPase signaling. Conversely, chronic administration of MBG to normotensive rats to achieve plasma concentrations of MBG as seen with PNx, produced cardiac phenotype similar to PNx.5,6

12 The transcription factor, Friend leukemia integration-1 (Fli-1), a member of the

ETS family, is a negative regulator of collagen synthesis,7 and reduced levels of Fli-1

were documented in skin fibroblasts of patients with scleroderma.8,9 Recent evidence

indicates that suppression of Fli-1 is also implicated in profibrotic signaling by CTS. In

vitro, we have demonstrated that nanomolar concentrations of MBG stimulate collagen

production by dermal, cardiac, and renal fibroblasts by a mechanism involving protein

kinase C δ-dependent phosphorylation and depletion of Fli-1.7 Interestingly, when we

stably transfected renal fibroblasts with a Fli-1 expression vector which dramatically increased Fli-1 expression, the basal expression of procollagen was decreased and MBG treatment did not increase procollagen expression or appreciably reduce Fli-1

expression.7

Recently, we developed two anti-MBG monoclonal antibodies (mAb), 3E9 and

4G4.10 In our previous experiments, 3E9 mAb exceeded 4G4 with respect to reversal of

MBG-induced Na/K ATPase inhibition, and potently reduced blood pressure and restored

vascular sodium pump activity in hypertensive Dahl-S rats and in pregnant Sprague-

Dawley rats rendered hypertensive by NaCl supplementation. Because of these

properties, in the present experiment we used 3E9 mAb for in vivo MBG

immunoneutralization, while 4G4 mAb which exhibits high affinity to MBG in

competitive immunoassays was chosen for MBG measurement.10 In the present

experiment, in PNx rats, we studied effects of 3E9 anti-MBG mAb on arterial pressure,

cardiac fibrosis and oxidative stress, and cardiac expression of Fli-1. We also compared

effects of 3E9 mAb to those of Digibind (the Fab fragments of ovine digoxin antibody)

which has been demonstrated to both bind endogenous CTS,11 as well as lower blood

13 pressure in patients with preeclampsia,12,13 a clinical syndrome known to have elevated

CTS levels.10,14

2.3 Methods

2.3.1 Animal studies

All animal experimentation described in this article was conducted in accordance

with the National Institutes of Health (NIH) Guide for the Care and Use of Laboratory

Animals under protocols approved by the University of Toledo Institutional Animal Care

and Use Committee. Male Sprague-Dawley rats (250–300 g) were used for these studies.

Eight sham nephrectomized rats comprised the control group. In 18 rats, PNx (5/6

nephrectomy) was produced by surgical removal of the right kidney and ligation of the

two-thirds of the arterial supply to the left kidney as reported previously in detail.15 In

brief, rats were anesthetized with a mixture of 100% oxygen and 5% isoflurane, an

incision was made in the left flank, through which the left kidney was pulled out, and

arteries supplying to upper and lower poles were ligated. After a week, the right kidney

was decapsulated to avoid removal of adrenal gland, artery, vein, and ureter were ligated,

and the kidney was removed. T his maneuver produces sustained hypertension within 2

weeks.5,6 At 4 weeks following PNx, these rats were intraperitoneally administered

vehicle (n = 6), Digibind (n = 6) or 3E9 anti-MBG mAb (n = 6). The dose of Digibind

(10 μg/kg) was similar to that previously administered to patients with preeclampsia,12,13

and the dose of 3E9 mAb (50 μg/kg) was the same as that previously reported to reverse

the EC75 to the inhibition of the Na/K-ATPase by MBG in rat renal outer medulla in

vitro, and to reduce blood pressure in hypertensive Dahl-S rats in vivo.10 Blood pressure

14 was determined using the tail cuff method by IITC (Amplifier model 229, Monitor model

31, Test chamber Model 306; IITC Life Science, Woodland Hills, CA) at baseline, 3, 24,

48 h and at 1 week following antibody treatment (5 weeks after PNx). Then rats were killed and the heart weight and cardiac histology were determined. Plasma samples were stored at −80 °C for determination of CTS.

2.3.2 Oxidative stress markers

Levels of oxidative stress were assessed by measurement of protein carbonyl levels and determination of intracellular production of reactive oxygen species using the fluorescent probe dye dihydroethidium (DHE). Total protein carbonyl concentration of the plasma and left ventricular homogenate was determined by enzyme-linked immunosorbent assay using the BIOCELL PC Test kit (Northwest Life Science

Specialties, Vancouver, WA). Production of reactive oxygen species was detected by

DHE (Invitrogen Molecular Probes, Eugene, OR) as described previously.16,17 Briefly, left ventricle tissue was frozen in optimal cutting temperature compound, and transverse sections (10 μm) were generated with a cryostat and placed on glass slides. Tissue sections were incubated with 5 μmol/l DHE at 37 °C for 20 min according to the manufacturer’s instructions. Red fluorescence was assessed by using an Olympus

FSX100 box type fluorescence imaging device (Olympus America, Center Valley, PA).

The excitation wavelength was 488 nm with emission at 585 nm. Fluorescence intensity was analyzed by the use of Image J (version 1.32j) software (National Institutes of

Health, USA; http://rsb.info.nih.gov/ij/).

15 2.3.3 Creatinine and creatinine clearance

At the conclusion of the study, 24 h urine samples were collected. At the end of

urine collection, animals were killed and blood samples were obtained from abdominal

aorta. Plasma creatinine was measured with a colorimetric method using a commercial kit from Teco Diagnostics (Anaheim, CA, cat. no. C515-480). Creatinine standards or plasma samples were mixed with the picric acid reagent and creatinine buffer reagent provided with the kit. The optical density value at 510 nm was measured immediately after and at 15 min. The differences between the two time points were used to calculate the creatinine concentrations. Creatinine clearance was calculated using the following formula: (urine Cr × urine Vol (ml)/plasma Cr × 24 h × 60 min).

2.3.4 Western blot analyses of Fli-1 and collagen-1

Western Blot analysis was performed on proteins from tissue homogenates as previously reported.7 The left ventricles from the heart were homogenized in ice-cold

RIPA lysis buffer (pH 7.0) Santa Cruz Biotechnology (Santa Cruz, CA; sc-24948). The

homogenate was centrifugated at 1,400g for 30 s at 4 °C. The supernatant was discarded

and the pellet fraction was resuspended in 5% sodium dodecyl sulfate (SDS) and 50

mmol/l Tris-HCl (pH 7.4). The protein was quantified in the resuspended pellet fraction

and was solubilized in sample buffer (2% SDS, 5% β-mercaptoethanol, 20% glycerol,

0.005% bromophenol blue, and 50mmol/l Tris- HCl; pH 7.0). The proteins, obtained

from tissue homogenates, were resolved on an SDS-polyacrylamide gel electrophoresis

(PAGE) using Precast Ready Gels 4–15% Tris-HCl, purchased from Bio-Rad (Hercules,

CA). Ten microgram of protein per sample were loaded into each well. The proteins

16 from the gel were electrotransferred to a nitrocellulose membrane. The membrane was

blocked with 5% nonfat dry milk in 20 mmol/l Tris-HCl (pH 7.5, 150 mmol/l NaCl, and

0.1% Tween 20). Goat anti-type 1 collagen antibody (Southern Biotech, Birmingham,

AL) was used to probe for collagen-1, and secondary anti-goat antibody was purchased

from Santa Cruz Biotechnology. To probe for Fli-1, we used rabbit polyclonal anti-

Fli1(C19) antibody (Santa Cruz Biotechnology; 1:500) and peroxidase-conjugated anti-

rabbit antiserum (Amersham, Piscataway, NJ; 1:1,000). For detection, we used ECL and

ECL plus purchased from Amersham Biosciences. Loading conditions were controlled using anti-actin mouse monoclonal antibody (Santa Cruz Biotechnology).

2.3.5 Histology

Trichrome staining was performed on left ventricular tissues and tissue fibrosis was quantified as previously reported.6–7,15 Left ventricle sections were immediately

fixed in 4% formalin buffer solution (pH 7.2) for 18 h, dehydrated in 70% ethanol, and

then embedded in paraffin and cut with a microtome. Trichrome staining was then

performed and fibrosis was quantified using ImageJ software. For quantitative

morphometric analysis, five random sections of trichrome slides were electronically

scanned into an RGB image which was subsequently analyzed using Image J (version

1.32j) software. The amount of fibrosis was then estimated from the RGB images with a

macro written by the authors (J.I.S.) by converting pixels of the image with substantially

greater (>120%) blue than red intensity to have the new, gray scale amplitude = 1,

leaving other pixels as with amplitude = 0.

17 2.3.6 MBG immunoassay

For measurement of MBG, plasma samples were extracted using C18 SepPak

cartridges (Waters, Cambridge, MA). Cartridges were activated with 10 ml acetonitrile and washed with 10 ml water. Then 0.5 ml plasma samples were applied to the cartridges and consecutively eluted in the same vial with 7 ml 20% acetonitrile followed by 7 ml

80% acetonitrile and vacuum dried. Before immunoassays, samples were reconstituted in the initial volume of assay buffer. MBG was measured using a fluoroimmunoassay based on a murine anti-MBG 4G4 mAb recently described in detail.10 This assay is based on competition between immobilized antigen (MBG-glycoside-thyroglobulin) and MBG or other cross-reactants, within the sample for a limited number of binding sites on 4G4 anti-MBG mAbs. Secondary (goat anti-mouse) antibody labeled with nonradioactive europium was obtained from Perkin-Elmer (Waltham, MA). Data on cross-reactivity of the 4G4 mAb used for determination of MBG levels and of 3E9 mAb used for in vivo administration was reported previously.10 MBG (>98% high performance liquid

chromatography pure) was purified from secretions from parotid glands of Bufo marinus

toads as reported previously.10

2.3.7 Statistical analyses

The results are presented as means ± s.e.m. Data were analyzed using one way analysis of variance followed by Newman–Keuls test (intragroup analyses), by repeated measures analysis of variance followed by Newman–Keuls test (intergroup analyses), and by two-tailed t-test (when applicable) (GraphPad Prism software, San Diego, CA). A two-sided P value of less than 0.05 was considered to be statistically significant.

18 2.4 Results

In Sprague-Dawley rats, PNx led to hypertension, marked increases in plasma creatinine and oxidative stress as assessed by plasma levels of carbonylated protein

(Table 2.1). Plasma levels of MBG in PNx rats were elevated fourfold vs. that in sham operated animals (Figure 2-1a). Rats subjected to PNx treated with vehicle developed cardiac hypertrophy (Figure 2-2a) which was accompanied by activation of cardiac oxidative stress assessed by carbonylated protein and DHE staining (Figure 2-2b, c).

Development of renal failure in rats was also associated with cardiac fibrosis assessed by computer-assisted morphological analysis and increased levels of collagen-1 in left ventricular myocardium (Figure 2-3) as we have previously reported.5,6 Cardiac levels of

Fli-1 in PNx rats were markedly reduced vs. that in sham-operated animals (Figure 2-3a).

Figures 2-1 and 2-2 summarize data on the effect of administration of 3E9 anti-

MBG mAb and Digibind on blood pressure, heart weight, and cardiac levels of carbonylated protein. A single administration of Digibind produced a transient decrease in arterial pressure while, in contrast, administration of 3E9 mAb resulted in a substantial and sustained decrease in systolic blood pressure following 1 week of antibody administration (Figure 2-1d). In addition to depressor effect, administration of 3E9 mAb and Digibind to PNx rats was associated with reduction in the serum levels of creatinine and a concomitant increase in creatinine clearance (Figure 2-1b, c).

Administration of both antibodies was associated with a reduction in cardiac weight and a decrease in the cardiac expression of carbonylated protein (Figure 2-2), as well as increase in the left ventricular expression of Fli-1 protein (Figure 2-3a) along with reductions in cardiac collagen 1 protein expression and morphological evidence of

19 fibrosis (Figure 2-3b, c). These effects of 3E9 mAb were more pronounced as compared

to those of Digibind.

2.5 Discussion

The main observation of the present experiment is that a single administration of a

mAb against an endogenous Na/KATPase inhibitor, MBG, to rats with experimental

renal failure produced a sustained depressor effect associated with a dramatic reduction in

cardiac fibrosis and increase in cardiac levels of Fli-1, a negative regulator of collagen

synthesis. Fli-1 belongs to a family of Ets oncogenes, and it competes with another

transcription factor, ETS-1, to maintain a balance between stimulation and repression of

Col1a2 gene promoter.8 Fli-1 is implicated in dermal fibrosis and it exhibits direct effect

on collagen-1 synthesis in dermal fibroblasts,8 and we have observed that decreases in

Fli-1 expression appear to be necessary for CTS to stimulate fibroblast collagen

production.7

Our present results demonstrate that blockade of the CTS-Na/ K-ATPase signal cascade can actually reverse established cardiac fibrosis in the PNx model. Our previous studies demonstrated that cardiac fibrosis was well established after 4 weeks following

PNx in the Sprague-Dawley rat.5,6 In the current study, treatment was administered to

rats at 4 weeks following PNx, and animals were killed 1 week later. While the vehicle

treatment group, studied 5 weeks after PNx, demonstrated similar degrees of cardiac

hypertrophy and fibrosis to what we had previously reported at 4 weeks,5,6 both Digibind

and 3E9 treated animals had remarkable clearing of myocardial fibrosis over the next

week accompanied by upregulation of cardiac Fli-1. These data further indicate that

20 MBG-dependent Fli-1 downregulation is implicated in the pathogenesis of cardiac

fibrosis seen with experimental chronic renal failure. Our present observations that

immunoneutralization of MBG was accompanied by reduction in systemic and cardiac

levels of oxidative stress agrees with previous data demonstrating that generation of

reactive oxygen species is implicated in MBG-dependent cell signaling.18,19

Previous studies in PNx rats implemented remnant kidney fibrosis in the

progression of renal failure in this model.20,21 Although absence of renal morphology

data is a limitation of the present study, we found that in PNx rats immunoneutralization

of CTS with both 3E9 mAb and Digibind reduced plasma creatinine concentration and

produced a substantial increase in creatinine clearance. This observation suggests that in

PNx rats beneficial effects of MBG immunoneutralization are not limited to pressor and

cardiac effects, and warrants further studies of the role of CTS in the pathogenesis of

renal fibrosis.

Notably, in the present study, in the case of Digibind, reduction of cardiac fibrosis

occurred in the absence of a sustained blood pressure-lowering effect. Thus, in the present experiment, Digibind and 3E9 mAb in PNx rats exhibited comparable antifibrotic effects in the presence of markedly varying effects on the blood pressure within 1 week after a single injection; while the blood pressure-lowering effect of anti-MBG mAb was profound and sustained, the depressor effect of Digibind was minor and transient. We, therefore, propose that in the present study, both antibodies exhibited blood pressure independent antifibrotic effects, which agrees with our previous data demonstrating that a pronounced antifibrotic effect of active immunization of PNx rats against MBG was associated with a very minor effect on the blood pressure.5,6

21 The 3E9 anti-MBG mAb employed in the current study is highly selective for

bufadienolide CTS and does not cross react with cardenolide sodium pump inhibitors and

other steroid hormones.10 Thus, in a competitive immunoassay, the 3E9 mAb exhibited

substantial cross-reactivity only with two , telocinobufagin, a possible precursor of MBG which was reported to be elevated in plasma of patients with uremia,22

and cinobufotalin which differs from MBG in having one extra hydroxyl group.10

Previously, we reported that following high-performance liquid chromatography-

fractionation of CTS from preeclamptic placentae, a competitive immunoassay based on

Digibind exhibited reactivity to high-performance liquid chromatography fractions having retention times similar to that seen with MBG and other bufadienolides, but not to ouabain-like immunoreactive material.23 Most recently, in patients with chronic kidney disease and in PNx rats we demonstrated that increase in plasma CTS detected by

Digibind is likely to reflect an increase in the levels of MBG.24 These observations

suggest that in renal failure MBG represents a target for Digibind.

In conclusion, in experimental chronic renal failure, elevated levels of MBG

contribute to hypertension and induce cardiac fibrosis via suppression of Fli-1,

representing a potential target for therapy. The effectiveness of 3E9 mAb for reversing

the cardiac disease in PNx animals and the fact that 3E9 mAb exhibits a long-lasting effect following a single injection, suggests a potential role for MBG immunoneutralization in patients with uremic cardiomyopathy.

22

2.6 Manuscript References

1. London GM. Cardiovascular disease in chronic renal failure: pathophysiologic

aspects. Semin Dial 2003; 16:85–94.

2. Remppis A, Ritz E. Cardiac problems in the dialysis patient: beyond coronary

disease. Semin Dial 2008; 21:319–325.

3. Bagrov AY, Shapiro JI. Endogenous digitalis: pathophysiologic roles and

therapeutic applications. Nat Clin Pract Nephrol 2008; 4:378–392.

4. Kometiani P, Li J, Gnudi L, Kahn BB, Askari A, Xie Z. Multiple signal transduction

pathways link Na+/K+-ATPase to growth-related genes in cardiac myocytes.

The roles of Ras and mitogen-activated protein kinases. J Biol Chem 1998;

273:15249–15256.

5. Kennedy DJ, Vetteth S, Periyasamy SM, Kanj M, Fedorova L, Khouri S,

Kahaleh MB, Xie Z, Malhotra D, Kolodkin NI, Lakatta EG, Fedorova OV,

Bagrov AY, Shapiro JI. Central role for the cardiotonic steroid marinobufagenin

in the pathogenesis of experimental uremic cardiomyopathy. Hypertension

2006; 47:488–495.

6. Elkareh J, Kennedy DJ, Yashaswi B, Vetteth S, Shidyak A, Kim EG, Smaili S,

Periyasamy SM, Hariri IM, Fedorova L, Liu J, Wu L, Kahaleh MB, Xie Z, Malhotra

D, Fedorova L, Kashkin VA, Bagrov AY, Shapiro JI. Marinobufagenin stimulates

fibroblast collagen production and causes fibrosis in experimental uremic

cardiomyopathy. Hypertension 2007;49:215-224.

23 7. Elkareh J, Periyasamy SM, Shidyak A, Vetteth S, Schroeder J, Raju V, Hariri IM,

El-Okdi N, Gupta S, Fedorova L, Liu J, Fedorova OV, Kahaleh MB, Xie Z, Malhotra

D, Watson DK, Bagrov AY, Shapiro JI. Marinobufagenin induces increases in

procollagen expression in a process involving protein kinase C and Fli-1:

implications for uremic cardiomyopathy. Am J Physiol Renal Physiol 2009;

296:F1219–F1226.

8. Czuwara-Ladykowska J, Shirasaki F, Jackers P, Watson DK, Trojanowska M. Fli-1

inhibits collagen type I production in dermal fibroblasts via an Sp1-dependent

pathway. J Biol Chem 2001; 276:20839–20848.

9. Wang Y, Fan PS, Kahaleh B. Association between enhanced type I collagen

expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts.

Arthritis Rheum 2006; 54:2271–2279.

10. Fedorova OV, Simbirtsev AS, Kolodkin NI, Kotov AY, Agalakova NI, Kashkin

VA, Tapilskaya NI, Bzhelyansky A, Reznik VA, Frolova EV, Nikitina ER, Budny

GV, Longo DL, Lakatta EG, Bagrov AY. Monoclonal antibody to an endogenous

bufadienolide, marinobufagenin, reverses preeclampsia-induced Na/K-ATPase

inhibition and lowers blood pressure in NaCl-sensitive hypertension. J Hypertens

2008; 26:2414–2425.

11. Fedorova OV, Tapilskaya NI, Bzhelyansky AM, Frolova EV, Nikitina ER,

Reznik VA, Kashkin VA, Bagrov AY. Interaction of Digibind with endogenous

cardiotonic steroids from preeclamptic placentae. J Hypertens 2010; 28:361–366.

24 12. Goodlin RC. Antidigoxin antibodies in eclampsia. N Engl J Med 1988; 318:518–

519.

13. Adair CD, Buckalew V, Taylor K, Ernest JM, Frye AH, Evans C, Veille JC.

Elevated endoxin-like factor complicating a multifetal second trimester pregnancy:

treatment with digoxin-binding immunoglobulin. Am J Nephrol 1996; 16:

529–531.

14. Lopatin DA, Ailamazian EK, Dmitrieva RI, Shpen VM, Fedorova OV, Doris PA,

Bagrov AY. Circulating bufodienolide and cardenolide sodium pump inhibitors in

preeclampsia. J Hypertens 1999; 17:1179–1187.

15. Shapiro JI, Harris DC, Schrier RW, Chan L. Attenuation of hypermetabolism in the

remnant kidney by dietary phosphate restriction in the rat. Am J Physiol 1990;

258:F183–F188.

16. Zanetti M, d’Uscio LV, Peterson TE, Katusic ZS, O’Brien T. Analysis of superoxide

anion production in tissue. Methods Mol Med 2005; 108:65–72.

17. Takimoto E, Champion HC, Li M, Ren S, Rodriguez ER, Tavazzi B, Lazzarino G,

Paolocci N, Gabrielson KL, Wang Y, Kass DA. Oxidant stress from nitric oxide

synthase-3 uncoupling stimulates cardiac pathologic remodeling from chronic

pressure load. J Clin Invest 2005; 115:1221–1231.

18. Xie Z, Kometiani P, Liu J, Li J, Shapiro JI, Askari A. Intracellular reactive oxygen

species mediate the linkage of Na+/K+-ATPase to hypertrophy and its marker

genes in cardiac myocytes. J Biol Chem 1999; 274:19323–19328.

25 19. Priyadarshi S, Valentine B, Han C, Fedorova OV, Bagrov AY, Liu J, Periyasamy

SM, Kennedy D, Malhotra D, Xie Z, Shapiro JI. Effect of green tea extract on

cardiachypertrophy following 5/6 nephrectomy in the rat. Kidney Int 2003; 63:1785–

1790.

20. An WS, Kim HJ, Cho KH, Vaziri ND. Omega-3 fatty acid supplementation

attenuates oxidative stress, inflammation, and tubulointerstitial fibrosis in

the remnant kidney. Am J Physiol Renal Physiol 2009; 297:

F895–F903.

21. Sun L, Zhang D, Liu F, Xiang X, Ling G, Xiao L, Liu Y, Zhu X, Zhan M, Yang Y,

Kondeti VK, Kanwar YS. Low-dose paclitaxel ameliorates fibrosis in the remnant

kidney model by down-regulating miR-192. J Pathol 2011; 225:364–377.

22. Komiyama Y, Dong XH, Nishimura N, Masaki H, Yoshika M, Masuda M,

Takahashi H. A novel endogenous digitalis, telocinobufagin, exhibits elevated plasma

levels in patients with terminal renal failure. Clin Biochem 2005; 38:36–45.

23. Fedorova OV, Tapilskaya NI, Bzhelyansky AM, Frolova EV, Nikitina ER,

Reznik VA, Kashkin VA, Bagrov AY. Interaction of Digibind with endogenous

cardiotonic steroids from preeclamptic placentae. J Hypertens 2010; 28:361–366.

24. Kolmakova EV, Haller ST, Kennedy DJ, Isachkina AN, Budny GV, Frolova EV,

Piecha G, Nikitina ER, Malhotra D, Fedorova OV, Shapiro JI, Bagrov AY.

Endogenous cardiotonic steroids in chronic renal failure. Nephrol Dial Transplant

2011; 26:2912–2919.

26 2.7 Table and Figure Legends

Table 2.1. Physiological measurements in control and PNx rats. Means ± SEM. *-

P<0.01 vs.control group. Two-tailed t-test or Wilcoxon test (plasma carbonylated protein). PNx, partially nephrectomized rats. *P<0.01 vs. control group.

Figure 2-1. Plasma levels of MBG (A), creatinine (B), and creatinine clearance (C)

in sham-operated (Sham) and PNx rats, treated with vehicle (Veh), Digibind (DG)

or anti-MBG mAb (3E9). Effects of administration of 3E9 anti-MBG mAb and of

Digibind to PNx rats on systolic BP (D). Means ± SEM from 6 observations. A: (*)

- P<0.01 vs. Sham by two-tailed t-test. B and C: (*) - P<0.05, (**) - P<0.001 vs.

Sham, (#) - P<0.05, (##) - P<0.01 vs. vehicle by one-way ANOVA followed by

Newman-Keuls test. D: By repeated measures ANOVA and Newman-Keuls test:

Digibind vs. vehicle – P<0.05; 3E9 mAb vs. vehicle – P<0.01; Digibind vs. 3E9 –

P<0.01.

Figure 2-2. Effects of administration of 3E9 anti-MBG mAb and of Digibind to PNx

rats on heart weight (A), and on cardiac levels of oxidative stress assessed by

measurement of carbonylated protein (B) and DHE fluorescence intensity (C);

upper panels, representative measurements; lower panel – quantitative

measurements, mean±SEM of 4 densiometry determinations). PNx – partially

nephrectomized rats. Vehicle – PNx rats administered vehicle. Means ± SEM

from 6 observations. A-C: By one-way ANOVA and Newman-Keuls test (*) –

27 P<0.05 and (**) – P<0.01 vs. Sham. (#) – P<0.05 and (##) – P<0.01 vs. vehicle treated

PNx rats.

Figure 2-3. Representative (upper panel) and quantitative (lower panel, mean±SEM of 4 densiometry measurements) analysis of Fli-1 (A) and collagen-1

(B) Western blots performed on cardiac tissues from the different groups. Actin

was used to control loading. C – representative (upper panel) and quantitative

(lower panel, mean±SEM of 4 densiometry measurements) trichrome-stained

photomicrographs obtained from cardiac tissue derived from the different

experimental groups. Sham – sham-operated rats, PNx – partially

nephrectomized rats, Veh – PNx rats administered vehicle, DG – PNx rats

administered Digibind, 3E9 – PNx rats administered 3E9 anti-MBG mAb. By one-way

ANOVA and Newman-Keuls test: (*) – P<0.01 vs. Sham; (#) – P<0.05, (##)

– P<0.01 vs. Veh.

28 2.8 Table and Figures

Table 2.1

29 Figure 2-1.

30 Figure 2-2.

31 Figure 2-3.

32 Chapter 3 – Manuscript

Title:

Rapamycin Reduces Cardiac Fibrosis in Experimental Uremic Cardiomyopathy

Authors:

Steven T. Haller1, George V. Budney1, Joe Xie1, Jiang Tian1, Mohamed Taleb1, Deepak Malhotra1, Olga V. Fedorova2, Alexei Y. Bagrov2, and Joseph I. Shapiro1

1University of Toledo Collage of Medicine, Toledo, Ohio

2 Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, Md.

Corresponding Author:

Dr. Joseph I. Shapiro, University of Toledo, Collage of Medicine, 3000 Arlington Ave., Toledo, OH

[email protected]

To be Submitted to Hypertension

33 3.1 Abstract

Background: We have shown that experimental uremic cardiomyopathy causes

cardiac fibrosis and is associated with increased levels of the cardiotonic steroid

marinobufagenin (MBG), an inhibitor of the Na/K-ATPase. The mammalian target of

rapamycin (mTOR) is a serine/threonine kinase implicated in the progression of many

different forms of renal disease. Treatment with rapamycin (an mTOR inhibor) has been

shown to attenuate inflammation, and renal fibrosis in experimental models of renal

disease. The use of rapamycin in the setting of experimental uremic cardiomyopathy has

not been defined.

Materials and Methods: Male Sprague Dawley rats weighing between 250-300

gms were used for these studies. Rats were divided into six groups. In the first group,

partial nephrectomy (PNx) was performed as we have previously described. This

maneuver produces sustained hypertension by 2 weeks under these conditions. In the

second group, PNx was performed and rapamycin was administered (0.2mg/kg/day). The

third group received both rapamycin (0.2mg/kg/day) and MBG (10µg/kg/day). The

fourth and fifth groups were administered MBG alone and rapamycin alone. The sixth

group consisted of sham operated controls. All treatments were performed for 4 weeks

with the use of osmotic minipumps.

Results: The PNx animals showed an extensive increase in plasma MBG levels,

systolic BP, and cardiac fibrosis. Plasma MBG levels were significantly decreased in the

PNx-rapamycin animals compared to PNx (124 ± 15 vs 342 ± 20, P<0.01). The PNx- rapamycin animals showed a substantial decrease in cardiac fibrosis compared to PNx

34

animals. MBG treated animals had significant increases in systolic BP, and cardiac fibrosis compared to controls. Rapamycin treatment in combination with MBG did not significantly attenuate these effects.

Conclusion: The mTOR pathway has been implicated in the generation of renal fibrosis during renal failure. Our results suggest that the mTOR pathway may have a significant impact in the generation of cardiac fibrosis. Treatment with rapamycin may provide a novel therapy for reducing cardiac fibrosis in the setting of uremic cardiomyopathy.

35

3.2 Introduction

The high mortality rate in patients with chronic renal failure is ultimately due to

severe cardiovascular disease. 1 This uremic cardiomyopathy is characterized by cardiac

hypertrophy, diastolic dysfunction, and cardiac fibrosis along with elevated circulating

concentrations of the cardiotonic steroid marinobufagenin (MBG), a ligand of the Na/K-

ATPase. MBG belongs to a family of bufadienolides previously described in

amphibians. 2 In toads, the biosynthesis of MBG occurs via the bile acid pathway from

cholic acids. 2 This pathway may also be responsible for the production of MBG in

mammals. We have shown that MBG is elevated in patients with renal failure 3 and in

rats subjected to partial nephrectomy (PNx), and those with pharmacologic administration of MBG developed a similar cardiomyopathy as seen in patients, whereas active immunization against MBG attenuated this in PNx. 4, 5 In PNx rats, we also

observed increased cardiac and plasma levels of carbonylated proteins as well as

evidence for signaling through the Na/K-ATPase such as activation of Src and mitogen-

activated protein kinase (MAPK).4, 5 A recent report from our group has shown that

treatment with a monoclonal antibody directed against MBG drastically reduced cardiac

fibrosis in PNx animals.6

The mammalian target of rapamycin (mTOR) is a serine/threonine kinase

composed of two signaling complexes, mTORC1 and mTORC2. 7 The mTORC1

complex is involved in cellular proliferation and growth, while mTORC2 is involved the

regulation of the cytoskeleton. 8 The mTOR pathway has been implicated in the

progression of many different forms of renal disease including experimentally induced

uremic cardiomyopathy .9, 10 Treatment with rapamycin (an mTORC1 inhibitor) has been 36

shown to attenuate inflammation, fibrosis, and cardiac hypertrophy in experimental

models of renal disease. 9, 10 Rapamycin is also a competitive inhibitor of CYP27A1, a

key rate limiting enzyme of the bile acid pathway .11

Based on this background, the primary goals of the present study were to

determine the effects of rapamycin on cardiac fibrosis and MBG production using the rat

PNx model of uremic cardiomyopathy.

3.3 Methods

3.3.1 Animal Studies

All animal experimentation described in this article was conducted in accordance with the National Institutes of Health (NIH) Guide for the Care and Use of Laboratory

Animals under protocols approved by the University of Toledo Institutional Animal Care and Use Committee. Male Sprague-Dawley rats (250–300 g) were used for these studies.

Rats were divided into six groups. In the first group, partial nephrectomy (PNx) was performed as we have previously described.5 In the second group, PNx was performed

and rapamycin (RAPA) was administered (0.2mg/kg/day). 12 The third group received

both rapamycin (0.2mg/kg/day) and MBG (10µg/kg/day). The fourth and fifth groups

were administered MBG alone (10µg/kg/day) and rapamycin alone (0.2mg/kg/day). The

dose of MBG is the same as we have previously reported to induce the physiological

changes associate with uremic cardiomyopathy .4, 5 The sixth group consisted of sham

37

operated controls. All treatments were performed for 4 weeks with the use of osmotic

minipumps (Alzet, model 2004). Minipumps were inserted SC through a flank incision.

3.3.2 Blood Pressure, Cardiac Physiology, and Other In Vivo Measurements

Blood pressure was determined using the tail cuff method by IITC (Amplifier

model 229, Monitor model 31, Test chamber Model 306; IITC Life Science, Woodland

Hills, CA) at baseline, and once weekly for four weeks. Then rats were euthanized and

the heart weight and cardiac histology were determined. Plasma samples were stored at

−80 °C for biochemical analysis. Plasma MBG and creatinine were measured as we have

previously described. 6, 13

3.3.3 Oxidative stress markers

Levels of oxidative stress were assessed by determination of intracellular production of reactive oxygen species using the fluorescent probe dye dihydroethidium

(DHE). Production of reactive oxygen species was detected by DHE (Invitrogen

Molecular Probes, Eugene, OR) as described previously. 14, 15 Briefly, left ventricle

tissue was frozen in optimal cutting temperature compound, and transverse sections (10

μm) were generated with a cryostat and placed on glass slides. Tissue sections were

incubated with 5 μmol/l DHE at 37 °C for 20 min according to the manufacturer’s

instructions. Red fluorescence was assessed by using an Olympus FSX100 box type

fluorescence imaging device (Olympus America, Center Valley, PA). The excitation

38

wavelength was 488 nm with emission at 585 nm. Fluorescence intensity was analyzed by the use of Image J (version 1.32j) software (National Institutes of Health, USA; http://rsb.info.nih.gov/ij/).

3.3.4 Isolation of Cardiac Fibroblasts and JEG-3 Cell Experiments

Isolation of cardiac fibroblasts was carried out as previously described by Brilla and coworkers 7 with modifications as previously reported.4 Briefly, male Sprague

Dawley rats weighing 250-300 grams were used to obtain fibroblast from the hearts. The rats were anesthetized with pentobarbital (50 mg/kg), and their hearts were removed and perfused under sterile condition via the ascending aorta with Joklik's medium (Sigma-

Aldrich, St. Louis, MO) in a modified Langendorffapparatus. After 5 min of perfusion, the perfusate was placed in Joklik's medium containing 0.1% collagenase type 2

(Worthington Biochemical, Lakewood, NJ) and0.1% BSA which was circulated for 15-

25 min until the heart became flaccid. Ventricles were excised and finely cut, and shaken in Joklik's modified medium with 0.1% collagenase and 0.1%BSA for15 min. Cells

/tissue suspension was allowed to settle for 15 min and was centrifugated at 500 rpm for

10 min. The supernatant then was centrifugated at 1500 rpm for 15 min. The resulting pellet was suspended in DMEM supplemented with antibiotics

(penicillin/streptomycin/fungizone) plus 15% FBS (Hyclone, Logan, UT) and seeded onto plates and incubated for 1hr. Unattached cells were removed, and the attached fibroblasts cells were allowed to grow until confluence and then trypsinized and passaged once at 1:3 dilution. Cells were allowed to grow confluent prior to use for experimental

39

purposes. All cells used in these experiments were from passage one unless otherwise

specified. Human placental chorionic epithelial cells (JEG-3) were purchased from a

commercially available vendor (ATCC), cultured in 6-well plates, and grown to confluence in minimum essential media over 48 hours. Cells were cultured in 2.5% FBS media and rapamycin (1uM) was added into half the wells. Both control and rapamycin- treated cells were sampled after 3, 6, and 12 hours of incubation. MBG was extracted from the collected media using C18 as reported.13 Competitive immunoassays were

performed using a monoclonal anti-MBG antibody to determine the concentration of

MBG in the samples.13

3.3.5 Western Blot Analysis

Western blot analysis was performed on proteins from tissue homogenates as

previously reported.4 For the cell lysates, the cells were grown to confluence and starved

for 18 h in DMEM with 1% FBS. The cells then were treated with MBG or rapamycin for

24 h when looking for procollagen expression. The cells were washed with phosphate

buffered saline (BPS) twice and exposed to lysis buffer. For detection of Collagen-1, the

left ventricles from the heart were homogenized in ice-cold RIPA lysis buffer (pH 7.0)

Santa Cruz Biotechnology (Santa Cruz, CA; sc-24948). The homogenate was

centrifugated at 1,400g for 30 s at 4 °C. The supernatant was discarded and the pellet

fraction was resuspended in 5% sodium dodecyl sulfate (SDS) and 50 mmol/l Tris-HCl

(pH 7.4). The protein was quantified in the resuspended pellet fraction and was

solubilized in sample buffer (2% SDS, 5% β-mercaptoethanol, 20% glycerol, 0.005%

40

bromophenol blue, and 50mmol/l Tris- HCl; pH 7.0). The proteins, obtained from tissue

homogenates, were resolved on an SDS-polyacrylamide gel electrophoresis (PAGE) using Precast Ready Gels 4–15% Tris-HCl, purchased from Bio-Rad (Hercules, CA).

Ten microgram of protein per sample were loaded into each well. The proteins from the gel were electrotransferred to a nitrocellulose membrane. The membrane was blocked with 5% nonfat dry milk in 20 mmol/l Tris-HCl (pH 7.5, 150 mmol/l NaCl, and 0.1%

Tween 20). Goat anti-type 1 collagen antibody (Southern Biotech, Birmingham, AL) was used to probe for collagen-1, and secondary anti-goat antibody was purchased from

Santa Cruz Biotechnology. For detection, we used ECL and ECL plus purchased from

Amersham Biosciences. Loading conditions were controlled using anti-actin mouse monoclonal antibody (Santa Cruz Biotechnology).

3.3.6 Histology

Left ventricle sections were immediately fixed in 4% formalin buffer solution

(pH 7.2) for 18 h, dehydrated in 70% ethanol, and then embedded in paraffin and cut with a microtome. Fast green staining with sirius red (0.1%) was performed on left ventricular tissues as described previously16, and fibrosis quantified using ImageJ

software (ImageJ 1.36b, National Institutes ofHealth, USA). For confirmation of the

histological findings, quantitative determination of Collagen-1 in left ventricular homogenates was performed using Western blot (as described above).

41

3.3.7 Statistical Analysis

The results are presented as means ± s.e.m. Data were analyzed using one way

analysis of variance followed by Newman–Keuls test (intragroup analyses), by repeated

measures analysis of variance followed by Newman–Keuls test (intergroup analyses), and

by two-tailed t-test (when applicable) (GraphPad Prism software, San Diego, CA). A

two-sided P value of less than 0.05 was considered to be statistically significant.

3.4 Results

3.4.1 Effect of Rapamycin on Blood Pressure, MBG Levels

Rapamycin treatment alone demonstrated a slight elevation in systolic BP, but did

not significantly alter MBG levels compared to control animals. PNx surgery

substantially increased the heart weight/body weight ratios. PNx surgery and MBG

infusion produced sustained hypertension throughout the duration of the experiment.

PNx MBG levels were similar to the MBG levels produced by MBG infusion alone. PNx

surgery with rapamycin infusion showed a significant decrease in systolic BP by the third

week which persisted at four weeks compared to PNx alone. The PNx surgery with

rapamycin treatment also demonstrated a drastic decrease in MBG levels.

Coadministration of MBG with rapamycin did not significantly attenuate systolic BP or

plasma MBG levels. These data are summarized in Table 3.1.

42

3.4.2 Effect of Rapamycin on Cardiac Fibrosis and oxidative stress

Cardiac fibrosis was assessed in the left ventricular myocardium by histological

analysis (sirius red with fast green staining) and collagen 1 expression determined by

Western blot. Both PNx and MBG infusion resulted in substantial increases of collagen

expression and cardiac scarring, while PNx with rapamycin infusion drastically lowered

these effects (Figure 3-1 A and B). Coadministration of rapamycin with MBG did not alter MBG induced cardiac fibrosis (Figure 3-1 A, and B). The increase in cardiac fibrosis in PNx and MBG treated animals was accompanied with an increase in cardiac oxidative stress as measured by DHE staining (Figure 3-2). Treatment with rapamycin did not alter oxidative stress in these animals.

3.4.3 Effect of Rapamycin on Fibroblast Procollagen Expression

Cultured cardiac fibroblasts treated with 1 and 100 nM of MBG resulted in a significant increase in procollagen 1 expression determined by Western blot (Figure 3.3).

Treatment with rapamycin (10.9 and 109 pM) significantly attenuated MBG (1 and 100 nM) induced procollagen 1 expression (Figure 3-3.) The concentrations of rapamycin chosen have been reported to have little effect on cell viability 17.

43

3.4.4 Effect of Rapamycin on MBG Production by JEG-3 Cells

Cultured human placental chorionic epithelial cells (JEG-3), which produce

MBG, were treated with 1 µM of rapamycin to test the effects on MBG production.

Rapamycin treatment (1 uM) at 3 and 6 hours significantly reduced MBG production compared to controls (84 vs 60 pmol/g protein, p<0.01; and 243 vs 116, pmol/g protein, p<0.01, Figure 3-4).

3.5 Discussion

The mTOR pathway has been shown to play a pivotal role in several different forms of renal disease.9 Treatment with rapamycin attenuates many of the physiological

changes associated with a decrease in renal function, including interstitial fibrosis.9 Our

current work demonstrates that rapamycin treatment in the setting of experimental uremic

cardiomyopathy significantly reduces cardiac fibrosis. This is in support of a recent

report with similar findings in a murine model of uremic cardiomyopathy.10 We have

also shown that fibroblast treatment with rapamycin drastically reduced procollagen

production in the presence of MBG. Importantly, rapamycin also drastically lowered

MBG levels in both the PNx model and in human placental chorionic epithelial cells

(JEG-3).

Recent work in animal models of renal disease has provided compelling evidence

for the involvement of mTORC1 in the generation of fibrosis. In an animal model of

unilateral obstructive nephropathy, as well as in fibroblasts, the profibrotic cytokine

44

TGF-β was shown to activate mTORC1 acting through a PI3K pathway. 18 In human

fibroblasts, the mTOR pathway has been shown to regulate collagen type I production. 19

Treatment with rapamycin has been shown to decrease TGF-β, fibroblast proliferation, and renal fibrosis. 18, 20, 21 In support of our results, rapamycin treatment significantly

decreased cardiac fibrosis as evaluated by trichrome staining in a murine model of uremic

cadiomyopathy.10 Similar results were reported using a transverse aortic constriction

model. 22

We have shown that MBG causes many of the pathophysiological changes

associated with experimental uremic cardiomyopathy including cardiac fibrosis 5, and

that MBG induces cardiac fibroblasts to produce collagen.4 The transcription factor

Friend leukemia integration-1 (Fli-1) acts as a negative regulator of collagen production

23, 24, and activation of protein kinase C-delta (PKC- δ) can phosphorylate Fli-1 to promote collagen synthesis. 25 We have recently reported that MBG induces

translocation of PKC-delta, which phosphorylates Fli-1 and leads to an increase in

collagen synthesis.26 Signaling through PKC-delta has been shown to activate the mTOR pathway.27

Treatment with rapamycin significantly reduced circulating MBG levels

compared to PNx animals. In addition, treatment with rapamycin in JEG-3 cells, which

produce MBG, resulted in a 52% reduction in MBG levels after six hours of treatment.

Endogenous cardiotonic steroids have been postulated to be synthesized from the classic

steroidogenesis pathway through cholesterol side-chain cleavage and pregnenolone precursors.2 Though this theory is still upheld for other cardiotonic steroids such as

45

ouabain, there have been controversial results with regard to MBG production.2 In toads, the biosynthesis of MBG occurs via the bile acid pathway form cholanic acids.2

Rapamycin acts as a competitive inhibitor of CYP27A1, a key rate-limiting enzyme of the bile acid pathway. Our data provides preliminary evidence indicating that the drastic reduction in MBG levels in both PNx animals and JEG-3 cells may be due to competitive inhibition of CYP27A1. Thus, in the setting of experimental uremic cardiomyopathy, rapamycin may have a duel effect of both inhibiting cardiac fibrosis and reducing MBG production.

Importantly, we did not see a significant reduction in cardiac fibrosis or MBG levels with combined MBG infusion and rapamycin treatment. We also did not see a reduction in oxidant stress in PNx animals treated with rapamycin or combined MBG and rapamycin treatment. We speculate that rapamycin was unable to provide a therapeutic effect in the setting of continuous MBG infusion. Furthermore, MBG induced ROS production may precede mTOR activation. Future experiments are warranted in order to determine if MBG is produced by the bile acid pathway in mammals, and if higher doses of rapamycin would overcome the effects of continuous MBG infusion.

In conclusion, the mTOR pathway has been implicated in the generation of renal fibrosis during renal failure. Our results suggest that the mTOR pathway may have a significant impact in the generation of cardiac fibrosis. Treatment with rapamycin may provide a novel therapy for reducing MBG levels and cardiac fibrosis in the setting of uremic cardiomyopathy.

46

3.6 Manuscript References 1. Sarnak MJ, Levey AS, Schoolwerth AC, Coresh J, Culleton B, Hamm LL,

McCullough PA, Kasiske BL, Kelepouris E, Klag MJ, Parfrey P, Pfeffer M, Raij

L, Spinosa DJ, Wilson PW. Kidney disease as a risk factor for development of

cardiovascular disease: a statement from the American Heart Association

Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research,

Clinical Cardiology, and Epidemiology and Prevention. Circulation.

2003;108(17):2154-2169.

2. Bagrov AY, Shapiro JI, Fedorova OV. Endogenous cardiotonic steroids:

physiology, pharmacology, and novel therapeutic targets. Pharmacol Rev.

2009;61(1):9-38.

3. Kolmakova EV, Haller ST, Kennedy DJ, Isachkina AN, Budny GV, Frolova EV,

Piecha G, Nikitina ER, Malhotra D, Fedorova OV, Shapiro JI, Bagrov AY.

Endogenous cardiotonic steroids in chronic renal failure. Nephrol Dial

Transplant.26(9):2912-2919.

4. Elkareh J, Kennedy DJ, Yashaswi B, Vetteth S, Shidyak A, Kim EG, Smaili S,

Periyasamy SM, Hariri IM, Fedorova L, Liu J, Wu L, Kahaleh MB, Xie Z,

Malhotra D, Fedorova OV, Kashkin VA, Bagrov AY, Shapiro JI.

Marinobufagenin stimulates fibroblast collagen production and causes fibrosis in

experimental uremic cardiomyopathy. Hypertension. 2007;49(1):215-224.

47

5. Kennedy DJ, Vetteth S, Periyasamy SM, Kanj M, Fedorova L, Khouri S, Kahaleh

MB, Xie Z, Malhotra D, Kolodkin NI, Lakatta EG, Fedorova OV, Bagrov AY,

Shapiro JI. Central role for the cardiotonic steroid marinobufagenin in the

pathogenesis of experimental uremic cardiomyopathy. Hypertension.

2006;47(3):488-495.

6. Haller ST, Kennedy DJ, Shidyak A, Budny GV, Malhotra D, Fedorova OV,

Shapiro JI, Bagrov AY. Monoclonal Antibody Against Marinobufagenin Reverses

Cardiac Fibrosis in Rats With Chronic Renal Failure. Am J Hypertens.

7. Hall MN. mTOR-what does it do? Transplant Proc. 2008;40(10 Suppl):S5-8.

8. Sarbassov DD, Ali SM, Kim DH, Guertin DA, Latek RR, Erdjument-Bromage H,

Tempst P, Sabatini DM. Rictor, a novel binding partner of mTOR, defines a

rapamycin-insensitive and raptor-independent pathway that regulates the

cytoskeleton. Curr Biol. 2004;14(14):1296-1302.

9. Lieberthal W, Levine JS. The role of the mammalian target of rapamycin (mTOR)

in renal disease. J Am Soc Nephrol. 2009;20(12):2493-2502.

10. Siedlecki AM, Jin X, Muslin AJ. Uremic cardiac hypertrophy is reversed by

rapamycin but not by lowering of blood pressure. Kidney Int. 2009;75(8):800-

808.

11. Gueguen Y, Ferrari L, Souidi M, Batt AM, Lutton C, Siest G, Visvikis S.

Compared effect of immunosuppressive drugs cyclosporine A and rapamycin on

cholesterol homeostasis key enzymes CYP27A1 and HMG-CoA reductase. Basic

Clin Pharmacol Toxicol. 2007;100(6):392-397.

48

12. Inman SR, Davis NA, Olson KM, Lukaszek VA, McKinley MR, Seminerio JL.

Rapamycin preserves renal function compared with cyclosporine A after

ischemia/reperfusion injury. Urology. 2003;62(4):750-754.

13. Fedorova OV, Simbirtsev AS, Kolodkin NI, Kotov AY, Agalakova NI, Kashkin

VA, Tapilskaya NI, Bzhelyansky A, Reznik VA, Frolova EV, Nikitina ER, Budny

GV, Longo DL, Lakatta EG, Bagrov AY. Monoclonal antibody to an endogenous

bufadienolide, marinobufagenin, reverses preeclampsia-induced Na/K-ATPase

inhibition and lowers blood pressure in NaCl-sensitive hypertension. J Hypertens.

2008;26(12):2414-2425.

14. Zanetti M, d'Uscio LV, Peterson TE, Katusic ZS, O'Brien T. Analysis of

superoxide anion production in tissue. Methods Mol Med. 2005;108:65-72.

15. Takimoto E, Champion HC, Li M, Ren S, Rodriguez ER, Tavazzi B, Lazzarino G,

Paolocci N, Gabrielson KL, Wang Y, Kass DA. Oxidant stress from nitric oxide

synthase-3 uncoupling stimulates cardiac pathologic remodeling from chronic

pressure load. J Clin Invest. 2005;115(5):1221-1231.

16. Lopez-De Leon A, Rojkind M. A simple micromethod for collagen and total

protein determination in formalin-fixed paraffin-embedded sections. J Histochem

Cytochem. 1985;33(8):737-743.

17. Poulalhon N, Farge D, Roos N, Tacheau C, Neuzillet C, Michel L, Mauviel A,

Verrecchia F. Modulation of collagen and MMP-1 gene expression in fibroblasts

by the immunosuppressive drug rapamycin. A direct role as an antifibrotic agent?

J Biol Chem. 2006;281(44):33045-33052.

49

18. Wang S, Wilkes MC, Leof EB, Hirschberg R. Noncanonical TGF-beta pathways,

mTORC1 and Abl, in renal interstitial fibrogenesis. Am J Physiol Renal

Physiol.298(1):F142-149.

19. Shegogue D, Trojanowska M. Mammalian target of rapamycin positively

regulates collagen type I production via a phosphatidylinositol 3-kinase-

independent pathway. J Biol Chem. 2004;279(22):23166-23175.

20. Shillingford JM, Piontek KB, Germino GG, Weimbs T. Rapamycin ameliorates

PKD resulting from conditional inactivation of Pkd1. J Am Soc

Nephrol.21(3):489-497.

21. Lloberas N, Cruzado JM, Franquesa M, Herrero-Fresneda I, Torras J, Alperovich

G, Rama I, Vidal A, Grinyo JM. Mammalian target of rapamycin pathway

blockade slows progression of diabetic kidney disease in rats. J Am Soc Nephrol.

2006;17(5):1395-1404.

22. Gao XM, Wong G, Wang B, Kiriazis H, Moore XL, Su YD, Dart A, Du XJ.

Inhibition of mTOR reduces chronic pressure-overload cardiac hypertrophy and

fibrosis. J Hypertens. 2006;24(8):1663-1670.

23. Czuwara-Ladykowska J, Shirasaki F, Jackers P, Watson DK, Trojanowska M. Fli-

1 inhibits collagen type I production in dermal fibroblasts via an Sp1-dependent

pathway. J Biol Chem. 2001;276(24):20839-20848.

24. Wang Y, Fan PS, Kahaleh B. Association between enhanced type I collagen

expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts.

Arthritis Rheum. 2006;54(7):2271-2279.

50

25. Jinnin M, Ihn H, Yamane K, Mimura Y, Asano Y, Tamaki K. Alpha2(I) collagen

gene regulation by protein kinase C signaling in human dermal fibroblasts.

Nucleic Acids Res. 2005;33(4):1337-1351.

26. Elkareh J, Periyasamy SM, Shidyak A, Vetteth S, Schroeder J, Raju V, Hariri IM,

El-Okdi N, Gupta S, Fedorova L, Liu J, Fedorova OV, Kahaleh MB, Xie Z,

Malhotra D, Watson DK, Bagrov AY, Shapiro JI. Marinobufagenin induces

increases in procollagen expression in a process involving protein kinase C and

Fli-1: implications for uremic cardiomyopathy. Am J Physiol Renal Physiol.

2009;296(5):F1219-1226.

27. Minhajuddin M, Bijli KM, Fazal F, Sassano A, Nakayama KI, Hay N, Platanias

LC, Rahman A. Protein kinase C-delta and phosphatidylinositol 3-kinase/Akt

activate mammalian target of rapamycin to modulate NF-kappaB activation and

intercellular adhesion molecule-1 (ICAM-1) expression in endothelial cells. J Biol

Chem. 2009;284(7):4052-4061.

51

3.7 Table and Figure Legends

Table 3.1. Effects of rapamycin on physiological measurements after PNx or infusion of MBG.

Sham refers to animals subject to sham surgery; PNx refers to partial nephrectomy; PNx

+ Rapa refers to PNx surgery and rapamycin infusion using minipumps; Rapa refers to rapamycin infusion using minipumps; MBG + Rapa refers to coadministration of MBG and rapamycin using minipumps; and MBG refers to MBG infusion using minipumps.*p<0.05 vs sham, †p<0.01 vs sham, §p<0.01 vs PNx, ‡p<0.01 vs MBG,

║p<0.01 vs PNx + Rapa, ¶p<0.01 vs MBG + Rapa

Figure 3-1. A, Representative (top) and quantitative analysis of collagen 1 (mean±SEM)

Western blots performed on cardiac tissue from the different groups. Actin was used as a loading control. B, Representative Sirius red and Fast green stained photomicrographs obtained from cardiac tissue derived from the different experimental groups. Amount of fibrosis expressed as mean±SEM measured using computer-assisted morphometry, as we have previously described 5. Sham refers to animals subject to sham surgery (n=8); PNx refers to partial nephrectomy (n=10); PNx + Rapa refers to PNx surgery and rapamycin infusion using minipumps (n=6); Rapa refers to rapamycin infusion using minipumps

(n=8); MBG + Rapa refers to coadministration of MBG and rapamycin using minipumps

(n=8); and MBG refers to MBG infusion using minipumps (n=8). *p<0.05 vs Sham,

**p<0.01 vs Sham, #p<0.01 vs PNx, ##p<0.01 vs Rapa, †p<0.05 vs Rapa.

52

Figure 3-2. Effects of rapamycin treatment on cardiac levels of oxidative stress assessed by DHE fluorescence intensity: (upper panels, representative measurements; lower panel

– quantitative measurements, mean±SEM of 4 densitometry determinations). Sham refers to animals subject to sham surgery; PNx refers to partial nephrectomy; PNx + Rapa refers to PNx surgery and rapamycin infusion using minipumps; Rapa refers to rapamycin infusion using minipumps; MBG + Rapa refers to coadministration of MBG and rapamycin using minipumps; and MBG refers to MBG infusion using minipumps.

*p<0.01 vs Sham, **p<0.01 vs Rapa.

Figure 3-3. Representative Western blot against procollagen 1 derived from cardiac fibroblasts treated with MBG (1 or 100 nM), rapamycin (10.9 or 109 pM), or a combination with the corresponding quantitative data shown as the mean±SEM of 5 experiments. *P<0.01 vs control, **p<0.01 vs MBG 100 nM, #P<0.01 vs MBG 1 nM

Figure 3-4. MBG production in JEG-3 cells after incubation with 1uM rapamycin for 3 and 6 hours. *p<0.05 vs control, **p<0.01 vs control.

53

3.8 Table and Figures

Table 3.1

54

Figure 3-1.

B Collagen-1

A Sham Rapa PNx Actin

PNx + Rapa MBG MBG + Rapa

55

Figure 3-2.

Sham Rapa PNx

PNx-Rapa MBG MBG-Rapa

56

Figure 3-3.

Procollagen-1

Actin

57

Figure 3-4.

58

Chapter 4 – Manuscript

Title:

Platelet Activation in Patients with Atherosclerotic Renal Artery Stenosis Undergoing Stent Revascularization

Authors:

Steven T. Haller1, MS, Satjit Adlakha1, D.O., Grant Reed2, M.D., Pamela Brewster1, MA, David Kennedy3, PhD, Mark W. Burket1, M.D., FACC, William Colyer1, M.D., Haifeng Yu1, MS, Dong Zhang1, MS, Joseph I. Shapiro1, MD, Christopher J. Cooper1, MD, FACC

1Department of Medicine, University of Toledo Collage of Medicine, Toledo, OH

2Brigham and Women’s Hospital, Boston, MA

3Cleveland Clinic Foundation, Cleveland, OH

Corresponding Author:

Steven T. Haller, University of Toledo, Collage of Medicine, 3000 Arlington Ave., Toledo, OH

[email protected]

Published in the Clinical Journal of the American Society of Nephrology, 2011 Sep;6(9):2185-91. Epub 2011 Aug 4.

59

4.1 Abstract

Background and Objectives: Soluble CD40 ligand (sCD40L) is a marker of platelet activation; whether platelet activation occurs in the setting of renal artery stenosis and stenting is unknown. Additionally, the effect of embolic protection devices and glycoprotein IIb/IIIa inhibitors on platelet activation during renal artery intervention is unknown.

Design, setting, participants, & measurements: Plasma levels of sCD40L were

measured in healthy controls, patients with atherosclerosis without renal stenosis, and

patients with renal artery stenosis before, immediately after, and 24hr after renal artery

stenting.

Results: Soluble CD40L levels were higher in renal artery stenosis patients than normal

controls (347.5 ± 27.0 vs 65.2 ± 1.4 pg/ml, p<0.001), but were similar to patients with

atherosclerosis without renal artery stenosis. Platelet rich emboli were captured in 26%

(9/35) of embolic protection device patients and in these patients sCD40L was elevated

prior to the procedure. Embolic protection device use was associated with a non-

significant increase in sCD40L whereas sCD40L declined with abciximab post-procedure

(324.9 ± 42.5 vs 188.7 ± 31.0 pg/ml, p=0.003) and at 24hrs.

Conclusions: Atherosclerotic renal artery stenosis is associated with platelet activation

but this appears to be related to atherosclerosis, not renal artery stenosis specifically.

Embolization of platelet rich thrombi is common in renal artery stenting and is inhibited

with abciximab.

60

4.2 Introduction

Platelet activation leading to thrombus formation is a well-described complication

of coronary artery disease, yet its occurrence in renal artery stenosis (RAS) is unknown. 1-

4 RAS is a major cause of secondary hypertension and an important cause of renal

failure.1-3, 5, 6 Although the utility of stent revascularization in patients with RAS is

uncertain, several studies suggest that at least a portion of patients develop a loss of

kidney function post-procedure. 1-3 6, 7

Soluble CD40 ligand (sCD40L) is expressed and secreted by platelets after

activation and plays a vital role in the immune, inflammatory, and coagulative responses

following injury or stress and in the setting of transplantation has been linked to renal

fibrosis .8-15 Moreover, high levels of sCD40L correlate with cardiovascular events in patients with unstable coronary syndromes. 13, 16-18 GP IIb/IIIa inhibitors may lower the

level of platelet activation in vitro and the level of sCD40L released from platelets upon

activation.19, 20 A recent report from our group has demonstrated that the use of a

GPIIb/IIIa inhibitor in combination with an embolic protection device (EPD) during renal

artery stenting may improve renal function following the revascularization procedure.21

However the relationship between platelet activation and patient outcome following renal artery stenting is uncertain.

On this background, the goals of the present study were to determine 1) if platelet activation is associated with atherosclerotic RAS, and 2) whether platelet activation occurs during renal artery stenting, and 3) if platelet thrombus formation captured by the

EPD correlates with systemic platelet activation .

61

4.3 Materials and Methods

The study, ClinicalTrials.gov identifier NCT00234585, was conducted with

funding provided by the sponsors, but study conduct, analysis, and reporting performed

independent of the sponsors. ICH good clinical practice guidelines were followed with

patients providing informed consent in an IRB approved protocol.

Platelet activation levels from the RAS patients were compared to 30 healthy

controls and, 30 patients with atherosclerosis undergoing coronary angiography, but free

of RAS. A total of 100 RAS patients were recruited from 7 sites. Inclusion required a

history of hypertension, renal insufficiency, heart failure, or angina with poorly

controlled hypertension and also the presence of 1 or more stenoses, ≥ 50% and <100, treatable with the EPD. RAS patients were randomized to the following allocations: 1/2 to Angioguard, 1/2 to no Angioguard; 1/2 to abciximab, 1/2 to placebo infusion yielding four groups: control, Angioguard only, abciximab only, and Angioguard with abciximab.

4.3.1 Pre-procedural care

In patients with RAS prior to double-blinded administration of Abciximab or placebo, systolic blood pressure was lowered to <160 mmHg. The target ACT was 275

seconds, if randomized to the EPD device an ACT of >300 seconds was required. A

bolus of 0.25 mg/kg abciximab (or placebo) was administered 5 minutes before crossing

the lesion, and was followed by an infusion at 0.125 µg/kg/min (maximum 10 µg /min) for 12 hours.

62

4.3.2 Central Laboratory Analysis

The blinded analysis of EPD contents was performed by the CV Path core lab

(Gaithersburg, MD). Platelet emboli consisted of layered platelet aggregates with

varying amounts of entrapped leukocytes and fibrin as evidence on hematoxylin and

eosin stained sections.22 Glomerular filtration rate (GFR), calculated from the modified

MDRD equation23, was used as the primary measure of renal function. Creatinine was

measured by a modified Jaffe reaction using the IDMS-traceable assay at the University

of Minnesota Core Lab for all subjects.

4.3.3 Blood Collection

Peripheral venous blood was collected at baseline, immediate post, and 24hr post procedure in lithium heparin plasma separator tubes, spun at 1000 x g for 15 minutes, and frozen at –80◦C until batch analysis.

4.3.4 Measurement of soluble CD40 ligand

Plasma levels of soluble CD40 ligand (sCD40L) were measured by enzyme linked immunosorbent assay (ELISA, R&D Systems; Minneapolis, MN). The ELISA kit

had intra-assay and interassay coefficients of 5% and 6%, respectively. The average

minimum detectible amount of sCD40L was 4.2 pg/ml.

63

4.3.5 Statistical Analysis

Study data are presented as continuous (mean±SEM) and categorical data.

Statistical analysis was performed on subjects with complete data for platelet activation measurements at the baseline, immediate post, and 24hr post procedure time points

(n=84). SAS one-way ANOVA were used to test for significance among groups.

Paired t-tests and Fisher’s PLSD post hoc tests were used to test for significance between groups. Unpaired t-tests were used to test for significance between the normal subjects, patient controls, and the RAS patients. Significance was defined as P<0.05. All analyses were performed in SAS or JMP.

4.4 Results

Baseline characteristics of the normal controls (n=30), atherosclerotic controls

(n=30), and the RAS patients (n=84), are shown in Table 4.1. The RAS patients had a significantly higher level of sCD40L compared to normal controls (347.5 ± 27.1 vs 65.2

± 1.4 pg/ml, P<0.001) (Figure 4-1). However, sCD40L levels were similar when compared to the patients with atherosclerosis who were free of renal artery stenosis

(347.5 ± 27.1 vs 335.2 ± 38.6 pg/ml, P=0.79) (Figure 4-1). Soluble CD40L, either at baseline or after the stenting, was not associated with baseline GFR or subsequent changes in kidney function.

64

4.4.1 EPD Content, Platelet Embolization, and sCD40L

Twenty six percent (9/35) of the patients that received the Angioguard had platelet rich emboli captured within the filter. In these patients with platelet rich emboli, sCD40L levels were higher than in patients without platelet emboli both before (497.9 ±

105.0 vs 313.7 ± 28.4 pg/ml, p=0.02) and after the procedure (443.3 ± 111.3 vs 232.2 ±

32.4 pg/ml, p=0.02) (Figure4-2)

4.4.2 Effect of Distal Protection and Drug Treatment

Patients with RAS randomized to abciximab had a significant decrease in

sCD40L levels immediately following the procedure (324.9 ± 42.5 vs 188.7 ± 31.0 pg/ml,

p=0.003), which persisted at 24hrs (324.9 ± 42.5 vs 181.2 ± 19.3 pg/ml, p=0.002) (Figure

4.3). In patients randomized to the Angioguard, sCD40L levels rose slightly immediately

following the procedure and at 24hrs (p=0.90) (Figure 4-3). Patients randomized to both

the Angioguard device and abciximab showed a significant decrease in sCD40L

immediately following the procedure (322.8 ± 35.2 vs 203.6 ± 33.1 pg/ml, p=0.03) but

this difference was no longer significant at 24 hours (Figure 4-3).

4.4.3 Effect of Abciximab and Clopidogrel

Clopiogrel use was not associated with lower sCD40L at baseline. For patients on

clopidogrel before intervention, sCD40L levels rose slightly immediately following the

procedure and decreased at 24hrs (p=0.53). For patients randomized to Abciximab and

not taking clopidogrel, sCD40L levels decreased significantly immediately following the

procedure (310.5 ± 33.0 vs 195.2 ± 31.3 pg/ml, p=0.008), which persisted at 24hrs (310.5 65

± 33.0 vs 173.1 ± 18.7 pg/ml, p<0.001. Patients on clopidogrel and randomized to

Abciximab showed a significant decrease in sCD40L at immediate post procedure (346.3

± 48.1 vs 198.3 ± 30.6, p=0.02). Similar effects were seen in patients that were

prescribed clopidogrel on the day of procedure.

4.5 Discussion

Platelet activation is a major cause of events and complications in coronary artery disease and with coronary revascularization. 24, 25 The use of platelet inhibitors during

coronary stenting reduces the potentially harmful effects of platelet activation including

abrupt vessel occlusion, myocardial infarction, and stent thrombosis. 25 To date, the

extent of platelet activation, and effect of anti-platelet therapies in the setting of renal

artery stenting has not been established. Thus, in the current study we sought to

determine whether atherosclerotic renal artery stenosis was associated with platelet

activation and the effect(s) of embolic protection and or use of platelet inhibitors on

markers of platelet activation.

Increased platelet activation is associated with a variety of vascular disorders

including acute coronary syndromes, stable coronary artery disease, and restenosis

following percutaneous coronary intervention. 26, 27 Soluble CD40L is a particularly

attractive marker for platelet activation since it is shed from the surface of activated

platelets, is easily measured, and meaningfully participates in a number of important

biologic processes including activation of immunity and thrombosis.28 The current study

found increased levels of sCD40L in the setting of RAS, however, this appears to be a

non-specific association with atherosclerosis in general as opposed to being attributable 66

to RAS specifically. More importantly though increased levels of sCD40L prior to the

procedure were more likely to have embolization of platelet-rich thrombi and these patients had persistently elevated levels of sCD40L after the procedure. This finding may represent a potentially modifiable feature denoting increased risk for patients referred for renal artery revascularization.

The current study also demonstrated that abciximab effectively inhibits platelet activation, as denoted by substantial suppression of sCD40L, up to 24 hrs following the procedure. Others have also observed the ability of GPIIb/IIIa inhibitors to lower levels of sCD40L in settings such as acute coronary syndromes and in STEMI patients undergoing coronary intervention. 29, 30 The current finding extends the prior observation that a GPIIb/IIIa inhibitor, when combined with an embolic protection device to capture atheroembolic debri, resulted in the most favorable renal function outcome. 21

The suppression of sCD40L release, observed with abciximab administration in

the current study, creates a plausible biologic pathway to explain the observation that

abciximab use was associated with improved renal function after stenting. In the kidney

the sCD40L/CD40 may be directly responsible for renal injury. Previously, others have

shown that angiotensin II stimulates release of renal TGF-β that in turn increases expression of the CD40 receptor on the proximal tubule of the kidney. 31 Pontrelli et al.

has shown that CD40 cross-linking on proximal tubular epithelial cells is pro-

inflammatory and induces fibrosis by stimulating the expression of plasminogen activator

inhibitor-1 (PAI-1) acting through a signaling pathway which is independent of the

proinflammatory signaling effects of CD40L.15 In addition, activation of the CD40

67

receptor results in infiltration of inflammatory cells into the interstitium of the kidney

through monocyte chemoattractant protein-1 (MCP-1) and intercellular adhesion molecule-1 (ICAM-1) expression.32 IL-8 amplifies CD40/CD154-mediated ICAM-1 production via the CXCR-1 receptor and p38-MAPK pathway in human renal proximal tubule cells.32 Furthermore, inhibition of the CD40/CD40L significantly decreased the

severity of renal injury in an animal model of chronic proteinuric renal disease.33 Thus, it

is conceivable that in patients with renal ischemia 1) the CD40 receptor is over-expressed

due to angiotensin II stimulation, 2) sCD40L shed by locally activated platelets may

activate the receptor and stimulate peritubular fibrosis in a manner independent of renal

blood flow or ischemia, and 3) this process may be accelerated at the time of a stent

procedure. In this regard the association between the GP IIb/IIIa inhibitor Abciximab

and improved renal function outcomes observed in the RESIST study21 may be

attributable to the drug’s effects in suppressing sCD40L as opposed to an effect on

thrombosis per se.

An observation from the RESIST study was that the EPD, when used without

abciximab, did not appear to improve renal function despite capturing debris. In the

current study we saw a slight increase in platelet activation with the use of the EPD

occurring immediately after the procedure, although this increase was not statistically

significant. Conceivably the EPD may slow blood flow in the vessel, provide a surface

upon which platelets can aggregate, and increase local platelet activation an effect

inhibited by the GPIIb/IIIa inhibitor. Admittedly the observed increase in circulating

levels of sCD40L with the use of the EPD was not statistically significant, however, it

68

may be unrealistic to expect that effects occurring on the surface of an EPD would be

detected systemically.

Several studies suggest a benefit of reducing platelet activation with loading doses

of 300-600 mg of clopidogrel prior to coronary interventions.34-36 However in the current

study pre-treatment with clopidogrel or clopidogrel administration on the day of

procedure did not significantly effect sCD40L levels. This may result from confounding

since patients were not randomized to clopidogrel treatment and had a significantly

higher prevalence of coronary artery and peripheral vascular disease, which may account

for the lack of difference observed in sCD40L levels. Work by Azar et al. reported a

reduction in sCD40L at a clopidogrel dose of 75 mg/day when preceded by a loading

dose of 300 mg in patients with stable CAD.37 Others though have failed to demonstrate

an effect of clopidogrel on levels of sCD40L.38

Increased levels of circulating sCD40L and the impact on renal function in the

setting of RAS remain speculative. Future clinical trials should address the effect of

sCD40L inhibition on distal embolization and renal function with long term follow-up.

The current study provides a foundation for exploring the role of CD40/CD40L signaling

and the generation of renal fibrosis during ischemic renal injury.

The following limitations of our study warrant mentioning: The current study

utilized sCD40L as the key measure of platelet activation. We did not measure sCD40L

at one month nor do we have longer-term follow-up of renal function beyond one month.

Thus, it remains uncertain whether other indices of platelet activation would provide

69

additional insights, or whether longer term follow up would have yielded similar results

for kidney function.

Atherosclerotic RAS is associated with increased platelet activation, but this increase appears to be attributable to atherosclerosis in general, not RAS specifically.

However, in patients with higher levels of platelet activation prior to the procedure, embolization of platelet rich thrombi is more common. Abciximab effectively inhibits platelet activation and sCD40L release, a mechanism that may explain the beneficial effect on renal function one month after the procedure that has been previously observed.

70

4.6 Manuscript References

1. Levin A, Linas S, Luft FC, Chapman AB, Textor S: Controversies in renal artery

stenosis: a review by the American Society of Nephrology Advisory Group on

Hypertension. Am J Nephro 27: 212-220. 2007

2. Balk E, Raman G, Chung M, Ip S, Tatsioni A, Alonso A, Chew P, Gilbert SJ, Lau

J: Effectiveness of management strategies for renal artery stenosis: a systematic

review. Ann Intern Med 145: 901-912. 2006

3. Olin JW: Survival in atherosclerotic renal artery stenosis: its all about renal

function, or is it? Catheter Cardiovasc Interv 69: 1048-1049. 2007

4. Arthurs Z, Starnes B, Cuadrado D, Sohn V, Cushner H, Andersen C: Renal artery

stenting slows the rate of renal function decline. J Vasc Surg 45: 726-731. 2007

5. Safian RD, Textor SC: Renal-artery stenosis. N Engl J Med 344: 431-442. 2001

6. Hirsch AT, Haskal ZJ, Hertzer NR, Bakal CW, Creager MA, Halperin JL,

Hiratzka LF, Murphy WR, Olin JW, Puschett JB, Rosenfield KA, Sacks D,

Stanley JC, Taylor LM, Jr., White CJ, White J, White RA, Antman EM, Smith

SC, Jr., Adams CD, Anderson JL, Faxon DP, Fuster V, Gibbons RJ, Hunt SA,

Jacobs AK, Nishimura R, Ornato JP, Page RL, Riegel B: ACC/AHA 2005

Practice Guidelines for the management of patients with peripheral arterial

disease (lower extremity, renal, mesenteric, and abdominal aortic): a collaborative

71

report from the American Association for Vascular Surgery/Society for Vascular

Surgery, Society for Cardiovascular Angiography and Interventions, Society for

Vascular Medicine and Biology, Society of Interventional Radiology, and the

ACC/AHA Task Force on Practice Guidelines (Writing Committee to Develop

Guidelines for the Management of Patients With Peripheral Arterial Disease):

endorsed by the American Association of Cardiovascular and Pulmonary

Rehabilitation; National Heart, Lung, and Blood Institute; Society for Vascular

Nursing; TransAtlantic Inter-Society Consensus; and Vascular Disease

Foundation. Circulation 113: e463-654. 2006

7. Cooper CJ, Murphy TP, Matsumoto A, Steffes M, Cohen DJ, Jaff M, Kuntz R,

Jamerson K, Reid D, Rosenfield K, Rundback J, D'Agostino R, Henrich W,

Dworkin L: Stent revascularization for the prevention of cardiovascular and renal

events among patients with renal artery stenosis and systolic hypertension:

rationale and design of the CORAL trial. Am Heart J 152: 59-66. 2006

8. Inwald DP, McDowall A, Peters MJ, Callard RE, Klein NJ: CD40 is

constitutively expressed on platelets and provides a novel mechanism for platelet

activation. Circ Res 92: 1041-1048. 2003

9. Chakrabarti S, Varghese S, Vitseva O, Tanriverdi K, Freedman JE: CD40 ligand

influences platelet release of reactive oxygen intermediates. Arterioscler Thromb

Vasc Biol 25: 2428-2434. 2005

72

10. Henn V, Slupsky JR, Grafe M, Anagnostopoulos I, Forster R, Muller-Berghaus G,

Kroczek RA: CD40 ligand on activated platelets triggers an inflammatory

reaction of endothelial cells. Nature 391: 591-594. 1998

11. Freedman JE: CD40-CD40L and platelet function: beyond hemostasis. Circ Res

92: 944-946. 2003

12. Andre P, Prasad KS, Denis CV, He M, Papalia JM, Hynes RO, Phillips DR,

Wagner DD: CD40L stabilizes arterial thrombi by a beta3 integrin--dependent

mechanism. Nat Med 8: 247-252. 2002

13. Mason PJ, Chakrabarti S, Albers AA, Rex S, Vitseva O, Varghese S, Freedman

JE: Plasma, serum, and platelet expression of CD40 ligand in adults with

cardiovascular disease. Am J Cardiol 96: 1365-1369. 2005

14. Santilli F, Davi G, Consoli A, Cipollone F, Mezzetti A, Falco A, Taraborelli T,

Devangelio E, Ciabattoni G, Basili S, Patrono C: Thromboxane-dependent CD40

ligand release in type 2 diabetes mellitus. J Am Coll Cardiol 47: 391-397. 2006

15. Pontrelli P, Ursi M, Ranieri E, Capobianco C, Schena FP, Gesualdo L,

Grandaliano G: CD40L proinflammatory and profibrotic effects on proximal

tubular epithelial cells: role of NF-kappaB and lyn. J Am Soc Nephrol 17: 627-

636. 2006

16. Nannizzi-Alaimo L, Rubenstein MH, Alves VL, Leong GY, Phillips DR, Gold

HK: Cardiopulmonary bypass induces release of soluble CD40 ligand.

Circulation 105: 2849-2854. 2002 73

17. Kritharides L, Lau GT, Freedman B: Soluble CD40 ligand in acute coronary

syndromes. N Engl J Med 348: 2575-2577. 2003

18. Burdon KP, Langefeld CD, Beck SR, Wagenknecht LE, Carr JJ, Rich SS,

Freedman BI, Herrington D, Bowden DW: Variants of the CD40 gene but not of

the CD40L gene are associated with coronary artery calcification in the Diabetes

Heart Study (DHS). Am Heart J 151: 706-711. 2006

19. Welt FG, Rogers SD, Zhang X, Ehlers R, Chen Z, Nannizzi-Alaimo L, Phillips

DR, Simon DI: GP IIb/IIIa inhibition with eptifibatide lowers levels of soluble

CD40L and RANTES after percutaneous coronary intervention. Catheter

Cardiovasc Interv 61: 185-189. 2004

20. Nannizzi-Alaimo L, Alves VL, Phillips DR: Inhibitory effects of glycoprotein

IIb/IIIa antagonists and aspirin on the release of soluble CD40 ligand during

platelet stimulation. Circulation 107: 1123-1128. 2003

21. Cooper CJ, Haller ST, Colyer W, Steffes M, Burket MW, Thomas WJ, Safian R,

Reddy B, Brewster P, Ankenbrandt MA, Virmani R, Dippel E, Rocha-Singh K,

Murphy TP, Kennedy DJ, Shapiro JI, D'Agostino RD, Pencina MJ, Khuder S:

Embolic protection and platelet inhibition during renal artery stenting. Circulation

117: 2752-2760. 2008

22. Burches B, Karnicki K, Wysokinski W, McBane RD, 2nd: Immunohistochemistry

of thrombi following iliac venous stenting: a novel model of venous thrombosis.

Thromb Haemost 96: 618-622. 2006

74

23. Stevens LA, Coresh J, Greene T, Levey AS: Assessing kidney function--

measured and estimated glomerular filtration rate. N Engl J Med 354: 2473-2483.

2006

24. Anand SX, Kim MC, Kamran M, Sharma SK, Kini AS, Fareed J, Hoppensteadt

DA, Carbon F, Cavusoglu E, Varon D, Viles-Gonzalez JF, Badimon JJ, Marmur

JD: Comparison of platelet function and morphology in patients undergoing

percutaneous coronary intervention receiving bivalirudin versus unfractionated

heparin versus clopidogrel pretreatment and bivalirudin. Am J Cardiol 100: 417-

424. 2007

25. Garg R, Uretsky BF, Lev EI: Anti-platelet and anti-thrombotic approaches in

patients undergoing percutaneous coronary intervention. Catheter Cardiovasc

Interv 70: 388-406. 2007

26. Santilli F, Basili S, Ferroni P, Davi G: CD40/CD40L system and vascular disease.

Intern Emerg Med 2: 256-268. 2007

27. Turker S, Guneri S, Akdeniz B, Ozcan MA, Baris N, Badak O, Kirimli O, Yuksel

F: Usefulness of preprocedural soluble CD40 ligand for predicting restenosis after

percutaneous coronary intervention in patients with stable coronary artery disease.

Am J Cardiol 97: 198-202. 2006

28. Antoniades C, Bakogiannis C, Tousoulis D, Antonopoulos AS, Stefanadis C: The

CD40/CD40 ligand system: linking inflammation with atherothrombosis. J Am

Coll Cardiol 54: 669-677. 2009

75

29. Furman MI, Krueger LA, Linden MD, Fox ML, Ball SP, Barnard MR, Frelinger

AL, 3rd, Michelson AD: GPIIb-IIIa antagonists reduce thromboinflammatory

processes in patients with acute coronary syndromes undergoing percutaneous

coronary intervention. J Thromb Haemost 3: 312-320. 2005

30. Dominguez-Rodriguez A, Abreu-Gonzalez P, Avanzas P, Bosa-Ojeda F, Samimi-

Fard S, Marrero-Rodriguez F, Kaski JC: Intracoronary versus intravenous

abciximab administration in patients with ST-elevation myocardial infarction

undergoing thrombus aspiration during primary percutaneous coronary

intervention--effects on soluble CD40 ligand concentrations. Atherosclerosis 206:

523-527. 2009

31. Starke A, Wuthrich RP, Waeckerle-Men Y: TGF-beta treatment modulates PD-L1

and CD40 expression in proximal renal tubular epithelial cells and enhances CD8

cytotoxic T-cell responses. Nephron Exp Nephrol 107: e22-29. 2007

32. Li H, Nord EP: IL-8 amplifies CD40/CD154-mediated ICAM-1 production via

the CXCR-1 receptor and p38-MAPK pathway in human renal proximal tubule

cells. Am J Physiol Renal Physiol 296: F438-445. 2009

33. Kairaitis L, Wang Y, Zheng L, Tay YC, Harris DC: Blockade of CD40-CD40

ligand protects against renal injury in chronic proteinuric renal disease. Kidney Int

64: 1265-1272. 2003

34. Nguyen TA, Lordkipanidze M, Diodati JG, Palisaitis DA, Schampaert E, Turgeon

J, Pharand C: Week-long high-maintenance dose clopidogrel regimen achieves

76

better platelet aggregation inhibition than a standard loading dose before

percutaneous coronary intervention: results of a double-blind, randomized clinical

trial. J Interv Cardiol 22: 368-377. 2009

35. Fefer P, Hod H, Hammerman H, Segev A, Beinart R, Boyko V, Behar S,

Matetzky S: Usefulness of pretreatment with high-dose clopidogrel in patients

undergoing primary angioplasty for ST-elevation myocardial infarction. Am J

Cardiol 104: 514-518. 2009

36. Gladding P, Webster M, Zeng I, Farrell H, Stewart J, Ruygrok P, Ormiston J, El-

Jack S, Armstrong G, Kay P, Scott D, Gunes A, Dahl ML: The antiplatelet effect

of higher loading and maintenance dose regimens of clopidogrel: the PRINC

(Plavix Response in Coronary Intervention) trial. JACC Cardiovasc Interv 1: 612-

619. 2008

37. Azar RR, Kassab R, Zoghbi A, Aboujaoude S, El-Osta H, Ghorra P, Germanos

M, Salame E: Effects of clopidogrel on soluble CD40 ligand and on high-

sensitivity C-reactive protein in patients with stable coronary artery disease. Am

Heart J 151: 521.e1-521.e4. 2006

38. Saw J, Madsen EH, Chan S, Maurer-Spurej E: The ELAPSE (Evaluation of Long-

Term Clopidogrel Antiplatelet and Systemic Anti-Inflammatory Effects) study. J

Am Coll Cardiol 52: 1826-1833. 2008

77

4.7 Table and Figure Legends

Table 4.1. Baseline Characteristics of the Normal Controls (Normal), Patient Controls

(Control), and the Renal Artery Stenosis Patients (RAS). Values are mean ± SD, or number and percentage of patients. BMI, body mass index; sCD40L, soluble CD40 ligand;

MI, myocardial infarction; NA, not applicable. aNormal vs. RAS. bPatient control vs. RAS

Figure 4-1. Soluble CD40L levels in Normal Controls, Patient Controls, and Renal

Artery Stenosis Patients (RAS). Box plot represents interquartile range with the median value shown as a horizontal bar within each box. Minimum and maximum values are shown in the bars outside each box. *p<0.001 vs Normal Control Subjects.

Figure 4-2. Soluble CD40L levels in patients With Platelet Rich Emboli Captured

Within the filter at Immediate Post Procedure. Analysis of Angioguard contents was performed in 35/39 (90%) of patients randomized to Angioguard. Nine (9) out of 35 patients (26%) had platelet-rich emboli captured. Data presented as mean ± SEM,

*p=0.02 vs Platelet Rich Emboli.

Figure 4-3. Soluble CD40L in patients with atherosclerotic renal artery stenosis randomly assigned to either abciximab, angioguard embolic protection, both, or neither.

Data presented as mean ± SEM, *p<0.01 vs Baseline, #p<0.05 vs Baseline.

78

4.8 Table and Figures

Table 4.1.

79

Figure 4-1.

700 *

* 600

500

400

sCD40L (pg/ml) sCD40L 300

200

100

0 Normal Control Patient Control RAS Patients Subjects (n=30) Subjects (n=30) (n=84)

80

Figure 4-2.

700.0

600.0

500.0

400.0 *

300.0 sCD40L (pg/ml) *

200.0

Platelet Rich Emboli (n=9) No Platelet Rich Emboli (n=26) 100.0

0.0 Baseline Immediate Post Procedure

81

Figure 4-3

82

Chapter 5 - CD40 Mediated Fibrosis in Chronic and Ischemic Renal Disease

5.1 Chronic Kidney Disease and Renal Ischemia

Recent data indicates that chronic kidney disease (CKD) is prevalent, affecting up

to 11% of the US adult population.1 Platelet activation and inflammation have been

implicated in the progression CKD.2 Cardiovascular disease is both common and a major

cause of mortality in patients with CKD.3, 4 This uremic cardiomyopathy is characterized

by a decrease in diastolic function, left ventricular hypertrophy, oxidant stress, and both

cardiac and renal fibrosis.5-7 We have shown that the cardiotonic steroid marinobufagenin

(MBG), signaling through the Na/K-ATPase, causes many of the adverse pathological effects of experimental uremic cardiomyopathy induced by 5/6th nephrectomy (PNx) in the rat. 8 CTS bind to the Na/K-ATPase and convert it into a signal transducer capable of activating multiple protein kinase cascades.9-11 Src binds to the Na/K-ATPase α1 subunit forming a functional signaling complex.12 CTS bind to the Na/K-ATPase and induce a conformational change which activates Src.12 Src transactivates EGFR which results in

the activation of phospholipase C (PLC), phosphoinsitide 3-kinase (PI3K), mitogen- activated protein kinases (MAPKs), protein kinase C (PKC), and the generation of reactive oxygen species (ROS).5, 11 We have demonstrated that pharmacologic

administration of MBG causes cardiac hypertrophy and fibrosis, as seen in patients,

83

whereas active immunization against MBG attenuated this in PNx.7, 8 Additionally, cardiac fibroblasts treated with MBG, at concentrations similar to those reported in experimental and clinical renal failure, has been shown to stimulate collagen production.7

This increase in collagen production appears to be dependent on the Na/K-ATPase-Src-

EGFR-ROS signaling cascade.7 The transcription factor Friend leukemia integration-1

(Fli-1) has been shown to be a negative regulator of collagen synthesis.13, 14 PKC- δ phosphorylates Fli-1 and promotes collagen synthesis.15 We have shown that MBG signaling through the Na/K-ATPase, cause PKC- δ translocation to the nucleus leading to

Fli-1 phosphorylation and collagen production.16

Renal artery stenosis (RAS) is a major cause of renal ischemia affecting 1-5% of the 60 million Americans with hypertension.17-19 Recent data suggests an incidence of up to 7% in patients over the age of 65.20 RAS is a major cause of secondary hypertension and an important cause of renal failure in patients with end stage renal disease. 21-24

Although the clinical utility of stent revascularization in patients with RAS is still uncertain, several studies suggest that at least a portion of patients develop a loss of kidney function post-procedure. 21, 22, 25, 26 Several mechanisms have been implicated as possible causes for a post-procedural decline in renal function such as contrast nephrotoxicity, and atheroembolization. Inflammation, fibrosis, and increases in oxidative stress leading to endothelial dysfunction have also been implicated in decreased renal function in the setting of atherosclerotic RAS.27 Increases in ROS have been reported in the stenotic kidneys from animal models of RAS.28, 29 We have shown that patients with RAS have significantly higher values of plasma MBG compared to healthy

84

control subjects, and patients with coronary atherosclerotic disease indicating that RAS

potentiates MBG release.30

5.2 Platelet Activation, CD40 Signaling, and Fibrosis

Increased platelet activation is associated with a variety of vascular disorders

including acute coronary syndromes, stable coronary artery disease, and restenosis

following percutaneous coronary intervention.31, 32 Platelet activation leads to crucial integrin mediated signaling cascades, which result in stable interactions between platelets and the endothelium as well as activation of glycoprotein IIb/IIIa receptors (primary aggregation receptors), release of alpha and dense granules, and expression and secretion of sCD40L.33, 34 Soluble CD40 ligand has been shown to play a vital role in the immune,

inflammatory, and coagulative responses following injury or stress, and implicated in the

generation of renal fibrosis.35-39 Moreover, high levels of sCD40L correlate with adverse cardiovascular events in patients with unstable coronary syndromes, including atherothrombotic lesions.35, 40-42

CD40, a type-I transmembrane receptor and a member of the tumor necrosis

factor (TNF) receptor superfamily 34, is expressed on a wide range of cells and critically

links thrombosis, inflammation, immunity, and fibrosis. Recent work in renal disease

models suggests that an important mediator of renal fibrosis and inflammatory cell

infiltration is CD40 that resides on the surface of the proximal tubular epithelium.

Specifically, CD40 is upregulated after renal injury 43 and activation of the receptor

85

results in 1) infiltration of inflammatory cells into the interstitium of the kidney through

monocyte chemoattractant protein-1 (MCP-1), and intracellular adhesion molecule-1

(ICAM-1) expression 44, and 2) increases plasminogen activator inhibitor type 1 (PAI-1)

and interstitial fibrosis.45-47 Importantly, Angiotensin II, whose release is increased

during renal ischemia, increases TGF-Β that in turn markedly increases expression of

CD40.47 Finally, CD40 activation increases antigen-specific recognition and killing of tubular epithelial cells by cytotoxic CD8+ T cells.47 Inhibition of CD40 significantly

decreased the severity of renal injury in an animal model of chronic proteinuric renal

disease.48 Our preliminary data shows a substantial increase in sCD40L in patients with

renal artery stenosis compared to normal control subjects, as well as increased expression

of CD40 in kidney tissue derived from PNx animals. We speculate that high levels of

systemic sCD40L travel distally to the kidney and activate CD40 on the proximal tubules

resulting in renal fibrosis.

5.3 Preliminary Data: Clinical trial

5.3.1 Soluble CD40 Ligand Levels in Patients with RAS

One hundred patients enrolled at 7 centers undergoing renal artery stenting were

randomized to an embolic protection device (EPD), or double-blind use of a GPIIb/IIIa inhibitor, Abciximab (the RESIST trial). A detailed description of the clinical trial has been previously reported.49 Plasma levels of sCD40L were measured in all patients with

available baseline blood samples (n=84) using a commercially available kit (R&D

86

Systems). Additional blood samples were collected from 30 healthy volunteers (Normal

Controls), and 30 patients with atherosclerosis, but without renal artery stenosis (Patient

Controls). A detailed description of this study has been reported and is described in

Chapter 4.50 Platelet activation is an important component of the atherosclerotic process

and shedding of sCD40L is a prominent feature of platelet activation. Our data confirms

that normal healthy controls, free of atherosclerosis, have low levels of circulating

sCD40L, whereas older subjects with atherosclerotic RAS are much higher (Figure 5-

1).50 Importantly, the increase in sCD40L is not specific to renal artery stenosis, since a

similar degree of elevation was observed in patients with atherosclerosis without RAS.

700 *

600 *

500

400

sCD40L (pg/ml) sCD40L 300

200

100

0 Normal Control Patient Control RAS Patients Subjects (n=30) Subjects (n=30) (n=84)

Figure 5-1: Soluble CD40 ligand levels in Normal Controls, Patient Controls, and RAS

Patients. *p<0.01 vs. Normal Controls.

87

5.3.2 Circulating concentrations of marinobufagenin (MBG) are substantially increased in patients with ischemic renal disease.

Plasma MBG levels were measured in patients with ischemic renal disease obtained from the aforementioned “RESIST” trial, non-RAS patient controls who were scheduled for coronary angiography, and normal healthy individuals. Marinobufagenin levels were noted to be significantly higher in patients with RAS compared with those of the other 2 groups (Figure 5-2).30 Multivariate analysis shows that occurrence of RAS is independently related to marinobufagenin levels. In addition, renal artery revascularization by stenting partially reversed marinobufagenin levels in the patients with RAS.30

Figure 5-2: MBG levels in Normal Controls, Patient Controls, and RAS Patients.

88

5.3.3 Soluble CD40 concentrations appear to predict outcomes in patients with

ischemic renal disease.

Very limited data is available on the effects of the CD40-sCD40L interactions on clinical outcome, with no published data in patients with ischemic RAS. Lajer et al reported higher levels of sCD40L in type-1 diabetics who developed nephropathy but no increase in mortality or rate of progression to ESRD.51 They had no data on CD40 and outcome. Recently we collaborated with Dr. Phillip Kalra in Manchester UK, assessing

CD40 and sCD40L (measured at the University of Toledo) in a single center cohort of

126 patients with atherosclerotic RAS followed longitudinally. We have observed a statistically significant inverse correlation between baseline circulating levels of CD40

(not tissue bound) and change in GFR over time. Specifically, there was less loss of GFR

(p=0.03) and better survival with higher baseline soluble CD40 (p=0.06). This is in agreement with the hypothesis that soluble CD40 “quenches” sCD40L thereby preventing activation of membrane-bound CD40. We also noted a trend toward increased mortality with higher levels of sCD40L (Figure 5-3).

89

45

40

35

30

25

20

Percent Mortality 15

10

5

0 < 100 sCD40L (pg/ml) 100-300 sCD40L (pg/ml) > 300 sCD40L (pg/ml)

Figure 5-3: Relationship between sCD40L and mortality rates in RAS patients. Note that in patients with high levels of sCD40L, mortality rates reach 40% during the follow up period obtained in the study. Because of the small numbers in this subset, the impressive trend is not statistically significant.

5.4 Preliminary Data: Animal Studies

We subjected Spargue-Dawley rats to either PNx surgery or infusion of MBG through minipumps at a dose designed to achieve similar elevations in plasma MBG as seen with PNx .8 We have previously demonstrated that this infusion of MBG produces substantial renal fibrosis by 4 weeks.6 When we examined the renal cortex of such animals (> 80% proximal tubule by volume), we noted substantial increases in CD40 expression (Figure 5-4, A). Of perhaps greater interest, we found that administration of either spironolactone or antibodies to MBG (either 3E9 mAb or digibind) resulted in marked decreases in renal fibrosis in the PNx model (Figure 5-5 and Figure 5-6, B). Our

90

work describing the use of 3E9 and digibind has recently been reported and is described

in detail in Chapter 2.52 We also noted marked increases in signaling through CD40 as

evidenced by PAI expression (Figure 5-4, B). We next observed that maneuvers which

prevented MBG signaling through the Na/K-ATPase, either administration of antibody

active against MBG or administration of the aldosterone antagonist, spironolactone,

which we have also demonstrated directly inhibits CTS binding to the Na/K-ATPase 53,

resulted in marked decreases in cortical CD40 expression (Figure 5-6, A, and Figure 5-7).

A B Sham PNx MBG Sham PNx MBG CD40 PAI-1

Actin Actin

3.5 2.00 1.80 3 * * 1.60 * ** 2.5 1.40 * 2 1.20

1.00 1.5 0.80 1 0.60

0.5 Expression1 of (Fraction Control) 0.40 - CD40 ExxpressionCD40 of (Fraction Control) PAI 0.20 0 Sham (n=10) PNx (n=10) MBG (n=8) 0.00 Sham (n=6) PNx (n=6) MBG (n=6)

Figure 5-4: Representative Western blot and quantitative data of (A) CD40 and (B) PAI-

1 expression derived form kidney cortex tissue, mean ± s.e.m. Sham (Sham operated

controls); PNx (5/6th nephrectomy); MBG (MBG infusion 10µg/kg/day). *p<0.01 vs.

Sham, **p<0.05 vs. PNx.

91

A B Collagen-1

Sham PNx 3E9 Dig

Sham PNx Actin

2.5 ** ** Dig 3E9 mAb 2 ## 7 * ** 6 # 1.5 5

4 Control) 1

* of (Fraction Expression 1

* - 3 # # 0.5 Fibrosis (% Area) 2 Collagen

1 0 0 Sham (n=6) PNx (n=6) 3E9 (n=6) Dig (n=6) Sham PNx DIG 3E9 mAb

Figure 5-5: Representative serius red fast green staining images (A), and (B) Western blot expression of collagen-1 with quantified data of kidney cortex tissue derived from

Sham, PNx, Digibind (Dig), and 3E9 mAb treated animals, mean ± s.e.m. **p<0.01 vs.

Sham, *p<0.05 vs. Sham, #p<0.01 vs. PNx, ##p<0.05 vs. PNx.

92

A Sham PNx PNx+SP SP B Sham PNx PNx+SP SP CD40 Collagen-1

Actin Actin 3.5 ** ## 3.5 ** 3 ## 3 2.5 ## * 2.5 2 # # 2 * 1.5 1.5 Control)

1 Expression1 of (Fraction - 1

0.5 0.5 CD40 ExpressionCD40 (Fraction of Control) Collagen 0 0 Sham (n=10) PNx (n=10) PNx+SP (n=8) SP (n=8) Sham (n=6) PNx (n=6) PNx + SP SP (n=6) (n=6) Figure 5-6: Representative Western blot and quantified data of (A) CD40, and (B) collagen-1 expression derived from kidney cortex tissue, mean ± s.e.m. Sham (Sham operated controls); PNx (5/6th nephrectomy); PNx+SP (PNx animals treated with spironolactone 80mg/kg/day); SP (Sham animals treated with spironolacton). **p<0.01 vs. Sham, *p<0.05 vs. Sham, #p<0.01 vs. PNx, ##p<0.01 vs. SP

93

Sham PNx 3E9 Dig

CD40

Actin

4 ** 3.5

3 ## * 2.5 * # 2

1.5

1

CD40 ExpressionCD40 (Fraction of Control) 0.5

0 Sham (n=7) PNx (n=6) 3E9 (n=6) Dig (n=6)

Figure 5-7: Representative Western blot and quatitiative data of CD40 expression derived fromkidney cortex tissue, mean ± s.e.m. Sham (Sham operated controls); PNx

(5/6th nephrectomy); 3E9 (PNx animals treated with 3E9 mAb); Dig (PNx animals treated with Digibind). **p<0.01 vs. Sham, *p<0.05 vs. Sham, #p<0.01 vs. PNx, ##p<0.05 vs.

PNx

5.5. Preliminary Data: LLC-PK1 cells

We have shown that MBG treatment in cardiac fibroblasts results in a substantial increase in procollagen-1 expression.7 Using a pig kiney proximal tubual cell line (LLC-

PK1 cells), we have demonstrated that MBG treatment resulted in a significant increase in both CD40 and procollagen-1 expression (Figure 5-8, A and B). Reactive oxygen 94

species (ROS) have been shown to induce CD40 signaling in vascular smooth muscle

cells.54 CTS signaling through the Na/K-ATPase induces ROS production and its

downsteam effects, such as cardiac and renal fibrosis, can be prevented by ROS

scavenging.5 Our collaborators have developed an LLC-PK1 cell line in which the alpha

1 isoform of the Na/K-ATPase (required for CTS signaling) has been knocked down

(PY-17 cells).55 Based on this background, we explored the potential crosstalk between

MBG induced ROS and CD40 expression using the previously described cell lines as

well as treatment with glucose oxidase (GO) which induces sustained levels of H2O2.

MBG and GO treatment resulted in a significant increase in CD40 and procollagen-1

expression, and this effect was midigated by treatment in PY-17 cells (Figures 5-9 and 5-

10). Admittedly, the decrease in procollagen-1 expression in PY-17 cells is more pronounced than the decrease in CD40 expression. Our in vivo results suggest that MBG signaling is required for procollagen-1 production, and contributes to increases in CD40 expression.

95

A B CD40 Procollagen-1

Actin Actin

2.5 3 * * 2.5 2

2 1.5 1.5

1 Control) 1

0.5 0.5 Procollagen Expression Expression of Procollagen (Fraction CD40 Expression (Fraction of Control) 0 0 Control (n=5) MBG 10nM (n=5) Control (n=5) MBG 10nM (n=5)

Figure 5-8: Representative Western blot and quantified data of CD40 expression (A) and procollagen-1 expression (B) from LLC-PK1 cells treated with MBG (10nM) for

24hrs (n=5 experiments). *

96

Control MBG (10nM) GO 1mU GO 3mU LLC PY-17 LLC PY-17 LLC PY-17 LLC PY-17 CD40

Actin

3 * * * # # # 2.5

2

1.5 CD 40 Expression (fraction (fraction control) of 1

0.5

0 LLC-Control PY-17 LLC-MBG PY-17-MBG LLC-GO 1mU PY-17-GO LLC-GO 3mU PY-17-GO Control (10nM) (10nM) 1mU 3mU

Figure 5-9: Representative Western blot and quantified data of CD40 expression from

LLC-PK1 cells (LLC) and PY-17 cells treated with MBG (10nM) and GO (1 and 3 mU) for 24hrs (n=5 experiments). *p<0.01 vs. Controls, #p<0.05 vs. PY-17-MBG, PY-17-GO

1mU, and PY-17-GO 3mU

97

Control MBG (10nM) GO 1mU GO 3mU LLC PY-17 LLC PY-17 LLC PY-17 LLC PY-17

Procollagen-1

Actin

* 3.5 # * * # 3 #

2.5

2

1.5 (fraction of of (fraction control) Procollagen Expression Expression Procollagen 1

0.5

0 LLC-Control PY-17 LLC-MBG PY-17-MBG LLC-GO 1mU PY-17-GO LLC-GO 3mU PY-17-GO Control (10nM) (10nM) 1mU 3mU

Figure 5-10: Representative Western blot and quantified data of procollagen-1 expression from LLC-PK1 cells (LLC) and PY-17 cells treated with MBG (10nM) and

GO (1 and 3 mU) for 24hrs (n=5 experiments). *p<0.01 vs. Controls, #p<0.01 vs. PY-17-

MBG, PY-17-GO 1mU, and PY-17-GO 3mU

98

5.6 Conclusions

The CD40/CD40L signaling cascade has been shown to induce inflammation and

fibrosis in proximal tubular epithelial cells.39 Specifically, stimulation of the CD40 receptor by sCD40L causes increased expression MCP-1 and the pro-fibrotic mediator

PAI-1 leading to the generation of fibrosis.39 Furthermore, inhibition of CD40/CD40L

signaling has been shown to decrease the severity of renal injury in an animal model of

chronic proteinuric renal disease.48 Our preliminary data from clinical trials demonstrates

that 1) plasma levels of sCD40L and MBG are significantly increased in patients with

ischemic renal disease, and 2) within these patients levels of circulation CD40 may

predict renal function. Our data in animal models suggests that PNx and MBG

administration results in significant increases in renal tissue expression of CD40 and PAI-

1 whereas treatment with spironolactone, and antibodies to inhibit CTS signaling

(digibind and 3E9) resulted in dramatic decreases in CD40 and collagen expression. Our

future work will focus on expanding our preliminary results in patients with ischemic

renal disease in order to determine if circulating levels of CD40 and CD40L are

predictive of renal function. In addition, we will use genetic manipulation both in vivo

and in vitro to knock out the CD40 receptor and knock down the Na/K-ATPase α-1

subunit. These manipulations will allow us to investigate the role of CTS signaling,

CD40 signaling, and the generation of renal fibrosis. Our ultimate goal is to provide

concrete evidence for our working hypothesis in the pathogenesis of renal fibrosis in the

setting of chronic and ischemic renal disease (Figure 5-11).

99

Figure 5-11: Potential scheme for CD40 mediate renal fibrosis in the setting of chronic and ischemic renal disease.

In the setting of chronic and ischemic renal injury, increased circulating levels of MBG convert the Na/K-ATPase into a signal transducer generating ROS, which may lead to increased expression of CD40 resulting in pro-inflammatory and pro-fibrotic signaling

cascades. CD40 signaling has also been implicated in the generation of ROS. Signaling

through the CD40 receptor could be stimulated by high circulating levels of sCD40L.

MBG signaling causes translocation of PKC-δ to the nucleus resulting in phosphorylation of FLI-1, and increased collagen expression leading to fibrosis.

100

5.7 References for Chapter 5

1. Kronenberg F. Emerging risk factors and markers of chronic kidney disease

progression. Nat Rev Nephrol. 2009;5(12):677-689.

2. Tu X, Chen X, Xie Y, Shi S, Wang J, Chen Y, Li J. Anti-inflammatory

renoprotective effect of clopidogrel and irbesartan in chronic renal injury. J Am

Soc Nephrol. 2008;19(1):77-83.

3. Chade AR, Lerman A, Lerman LO. Kidney in early atherosclerosis.

Hypertension. 2005;45(6):1042-1049.

4. Jaradat MI, Molitoris BA. Cardiovascular disease in patients with chronic kidney

disease. Semin Nephrol. 2002;22(6):459-473.

5. Bagrov AY, Shapiro JI, Fedorova OV. Endogenous cardiotonic steroids:

physiology, pharmacology, and novel therapeutic targets. Pharmacol Rev.

2009;61(1):9-38.

6. Fedorova LV, Raju V, El-Okdi N, Shidyak A, Kennedy DJ, Vetteth S,

Giovannucci DR, Bagrov AY, Fedorova OV, Shapiro JI, Malhotra D. The

cardiotonic steroid hormone marinobufagenin induces renal fibrosis: implication

of epithelial-to-mesenchymal transition. Am J Physiol Renal Physiol.

2009;296(4):F922-934.

7. Elkareh J, Kennedy DJ, Yashaswi B, Vetteth S, Shidyak A, Kim EG, Smaili S,

Periyasamy SM, Hariri IM, Fedorova L, Liu J, Wu L, Kahaleh MB, Xie Z,

Malhotra D, Fedorova OV, Kashkin VA, Bagrov AY, Shapiro JI.

101

Marinobufagenin stimulates fibroblast collagen production and causes fibrosis in

experimental uremic cardiomyopathy. Hypertension. 2007;49(1):215-224.

8. Kennedy DJ, Vetteth S, Periyasamy SM, Kanj M, Fedorova L, Khouri S, Kahaleh

MB, Xie Z, Malhotra D, Kolodkin NI, Lakatta EG, Fedorova OV, Bagrov AY,

Shapiro JI. Central role for the cardiotonic steroid marinobufagenin in the

pathogenesis of experimental uremic cardiomyopathy. Hypertension.

2006;47(3):488-495.

9. Xie Z, Askari A. Na(+)/K(+)-ATPase as a signal transducer. Eur J Biochem.

2002;269(10):2434-2439.

10. Xie Z, Cai T. Na+-K+--ATPase-mediated signal transduction: from protein

interaction to cellular function. Mol Interv. 2003;3(3):157-168.

11. Xie Z. Molecular mechanisms of Na/K-ATPase-mediated signal transduction.

Ann N Y Acad Sci. 2003;986:497-503.

12. Tian J, Cai T, Yuan Z, Wang H, Liu L, Haas M, Maksimova E, Huang XY, Xie

ZJ. Binding of Src to Na+/K+-ATPase forms a functional signaling complex. Mol

Biol Cell. 2006;17(1):317-326.

13. Czuwara-Ladykowska J, Shirasaki F, Jackers P, Watson DK, Trojanowska M. Fli-

1 inhibits collagen type I production in dermal fibroblasts via an Sp1-dependent

pathway. J Biol Chem. 2001;276(24):20839-20848.

14. Wang Y, Fan PS, Kahaleh B. Association between enhanced type I collagen

expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts.

Arthritis Rheum. 2006;54(7):2271-2279.

102

15. Jinnin M, Ihn H, Yamane K, Mimura Y, Asano Y, Tamaki K. Alpha2(I) collagen

gene regulation by protein kinase C signaling in human dermal fibroblasts.

Nucleic Acids Res. 2005;33(4):1337-1351.

16. Elkareh J, Periyasamy SM, Shidyak A, Vetteth S, Schroeder J, Raju V, Hariri IM,

El-Okdi N, Gupta S, Fedorova L, Liu J, Fedorova OV, Kahaleh MB, Xie Z,

Malhotra D, Watson DK, Bagrov AY, Shapiro JI. Marinobufagenin induces

increases in procollagen expression in a process involving protein kinase C and

Fli-1: implications for uremic cardiomyopathy. Am J Physiol Renal Physiol.

2009;296(5):F1219-1226.

17. Derkx FH, Schalekamp MA. Renal artery stenosis and hypertension. Lancet.

1994;344(8917):237-239.

18. Ram CV. Renovascular hypertension. Cardiol Clin. 1988;6(4):483-508.

19. Vokonas PS, Kannel WB, Cupples LA. Epidemiology and risk of hypertension in

the elderly: the Framingham Study. J Hypertens Suppl. 1988;6(1):S3-9.

20. Hansen KJ, Edwards MS, Craven TE, Cherr GS, Jackson SA, Appel RG, Burke

GL, Dean RH. Prevalence of renovascular disease in the elderly: a population-

based study. J Vasc Surg. 2002;36(3):443-451.

21. Balk E, Raman G, Chung M, Ip S, Tatsioni A, Alonso A, Chew P, Gilbert SJ, Lau

J. Effectiveness of management strategies for renal artery stenosis: a systematic

review. Ann Intern Med. 2006;145(12):901-912.

22. Olin JW. Survival in atherosclerotic renal artery stenosis: its all about renal

function, or is it? Catheter Cardiovasc Interv. 2007;69(7):1048-1049.

103

23. Safian RD, Textor SC. Renal-artery stenosis. N Engl J Med. 2001;344(6):431-

442.

24. Hirsch AT, Haskal ZJ, Hertzer NR, Bakal CW, Creager MA, Halperin JL,

Hiratzka LF, Murphy WR, Olin JW, Puschett JB, Rosenfield KA, Sacks D,

Stanley JC, Taylor LM, Jr., White CJ, White J, White RA, Antman EM, Smith

SC, Jr., Adams CD, Anderson JL, Faxon DP, Fuster V, Gibbons RJ, Hunt SA,

Jacobs AK, Nishimura R, Ornato JP, Page RL, Riegel B. ACC/AHA 2005

Practice Guidelines for the management of patients with peripheral arterial

disease (lower extremity, renal, mesenteric, and abdominal aortic): a collaborative

report from the American Association for Vascular Surgery/Society for Vascular

Surgery, Society for Cardiovascular Angiography and Interventions, Society for

Vascular Medicine and Biology, Society of Interventional Radiology, and the

ACC/AHA Task Force on Practice Guidelines (Writing Committee to Develop

Guidelines for the Management of Patients With Peripheral Arterial Disease):

endorsed by the American Association of Cardiovascular and Pulmonary

Rehabilitation; National Heart, Lung, and Blood Institute; Society for Vascular

Nursing; TransAtlantic Inter-Society Consensus; and Vascular Disease

Foundation. Circulation. 2006;113(11):e463-654.

25. Levin A, Linas S, Luft FC, Chapman AB, Textor S. Controversies in renal artery

stenosis: a review by the American Society of Nephrology Advisory Group on

Hypertension. Am J Nephrol. 2007;27(2):212-220.

26. Cooper CJ, Murphy TP, Matsumoto A, Steffes M, Cohen DJ, Jaff M, Kuntz R,

Jamerson K, Reid D, Rosenfield K, Rundback J, D'Agostino R, Henrich W,

104

Dworkin L. Stent revascularization for the prevention of cardiovascular and renal

events among patients with renal artery stenosis and systolic hypertension:

rationale and design of the CORAL trial. Am Heart J. 2006;152(1):59-66.

27. Lerman LO, Textor SC, Grande JP. Mechanisms of tissue injury in renal artery

stenosis: ischemia and beyond. Prog Cardiovasc Dis. 2009;52(3):196-203.

28. Chade AR, Rodriguez-Porcel M, Herrmann J, Zhu X, Grande JP, Napoli C,

Lerman A, Lerman LO. Antioxidant intervention blunts renal injury in

experimental renovascular disease. J Am Soc Nephrol. 2004;15(4):958-966.

29. Zhu XY, Chade AR, Rodriguez-Porcel M, Bentley MD, Ritman EL, Lerman A,

Lerman LO. Cortical microvascular remodeling in the stenotic kidney: role of

increased oxidative stress. Arterioscler Thromb Vasc Biol. 2004;24(10):1854-

1859.

30. Tian J, Haller S, Periyasamy S, Brewster P, Zhang H, Adlakha S, Fedorova OV,

Xie ZJ, Bagrov AY, Shapiro JI, Cooper CJ. Renal ischemia regulates

marinobufagenin release in humans. Hypertension.56(5):914-919.

31. Santilli F, Basili S, Ferroni P, Davi G. CD40/CD40L system and vascular disease.

Intern Emerg Med. 2007;2(4):256-268.

32. Turker S, Guneri S, Akdeniz B, Ozcan MA, Baris N, Badak O, Kirimli O, Yuksel

F. Usefulness of preprocedural soluble CD40 ligand for predicting restenosis after

percutaneous coronary intervention in patients with stable coronary artery disease.

Am J Cardiol. 2006;97(2):198-202.

33. Davi G, Patrono C. Platelet activation and atherothrombosis. N Engl J Med.

2007;357(24):2482-2494.

105

34. Antoniades C, Bakogiannis C, Tousoulis D, Antonopoulos AS, Stefanadis C. The

CD40/CD40 ligand system: linking inflammation with atherothrombosis. J Am

Coll Cardiol. 2009;54(8):669-677.

35. Mason PJ, Chakrabarti S, Albers AA, Rex S, Vitseva O, Varghese S, Freedman

JE. Plasma, serum, and platelet expression of CD40 ligand in adults with

cardiovascular disease. Am J Cardiol. 2005;96(10):1365-1369.

36. Inwald DP, McDowall A, Peters MJ, Callard RE, Klein NJ. CD40 is

constitutively expressed on platelets and provides a novel mechanism for platelet

activation. Circ Res. 2003;92(9):1041-1048.

37. Chakrabarti S, Varghese S, Vitseva O, Tanriverdi K, Freedman JE. CD40 ligand

influences platelet release of reactive oxygen intermediates. Arterioscler Thromb

Vasc Biol. 2005;25(11):2428-2434.

38. Henn V, Slupsky JR, Grafe M, Anagnostopoulos I, Forster R, Muller-Berghaus G,

Kroczek RA. CD40 ligand on activated platelets triggers an inflammatory

reaction of endothelial cells. Nature. 1998;391(6667):591-594.

39. Pontrelli P, Ursi M, Ranieri E, Capobianco C, Schena FP, Gesualdo L,

Grandaliano G. CD40L proinflammatory and profibrotic effects on proximal

tubular epithelial cells: role of NF-kappaB and lyn. J Am Soc Nephrol.

2006;17(3):627-636.

40. Nannizzi-Alaimo L, Rubenstein MH, Alves VL, Leong GY, Phillips DR, Gold

HK. Cardiopulmonary bypass induces release of soluble CD40 ligand.

Circulation. 2002;105(24):2849-2854.

106

41. Kritharides L, Lau GT, Freedman B. Soluble CD40 ligand in acute coronary

syndromes. N Engl J Med. 2003;348(25):2575-2577; author reply 2575-2577.

42. Burdon KP, Langefeld CD, Beck SR, Wagenknecht LE, Carr JJ, Rich SS,

Freedman BI, Herrington D, Bowden DW. Variants of the CD40 gene but not of

the CD40L gene are associated with coronary artery calcification in the Diabetes

Heart Study (DHS). Am Heart J. 2006;151(3):706-711.

43. Gaweco AS, Mitchell BL, Lucas BA, McClatchey KD, Van Thiel DH. CD40

expression on graft infiltrates and parenchymal CD154 (CD40L) induction in

human chronic renal allograft rejection. Kidney Int. 1999;55(4):1543-1552.

44. Li H, Nord EP. IL-8 amplifies CD40/CD154-mediated ICAM-1 production via

the CXCR-1 receptor and p38-MAPK pathway in human renal proximal tubule

cells. Am J Physiol Renal Physiol. 2009;296(2):F438-445.

45. Pontrelli P, Rossini M, Infante B, Stallone G, Schena A, Loverre A, Ursi M,

Verrienti R, Maiorano A, Zaza G, Ranieri E, Gesualdo L, Ditonno P, Bettocchi C,

Schena FP, Grandaliano G. Rapamycin inhibits PAI-1 expression and reduces

interstitial fibrosis and glomerulosclerosis in chronic allograft nephropathy.

Transplantation. 2008;85(1):125-134.

46. Rerolle JP, Hertig A, Nguyen G, Sraer JD, Rondeau EP. Plasminogen activator

inhibitor type 1 is a potential target in renal fibrogenesis. Kidney Int.

2000;58(5):1841-1850.

47. Starke A, Wuthrich RP, Waeckerle-Men Y. TGF-beta treatment modulates PD-L1

and CD40 expression in proximal renal tubular epithelial cells and enhances CD8

cytotoxic T-cell responses. Nephron Exp Nephrol. 2007;107(1):e22-29.

107

48. Kairaitis L, Wang Y, Zheng L, Tay YC, Harris DC. Blockade of CD40-CD40

ligand protects against renal injury in chronic proteinuric renal disease. Kidney

Int. 2003;64(4):1265-1272.

49. Cooper CJ, Haller ST, Colyer W, Steffes M, Burket MW, Thomas WJ, Safian R,

Reddy B, Brewster P, Ankenbrandt MA, Virmani R, Dippel E, Rocha-Singh K,

Murphy TP, Kennedy DJ, Shapiro JI, D'Agostino RD, Pencina MJ, Khuder S.

Embolic protection and platelet inhibition during renal artery stenting.

Circulation. 2008;117(21):2752-2760.

50. Haller S, Adlakha S, Reed G, Brewster P, Kennedy D, Burket MW, Colyer W, Yu

H, Zhang D, Shapiro JI, Cooper CJ. Platelet activation in patients with

atherosclerotic renal artery stenosis undergoing stent revascularization. Clin J Am

Soc Nephrol.6(9):2185-2191.

51. Lajer M, Tarnow I, Michelson AD, Jorsal A, Frelinger AL, Parving HH, Rossing

P, Tarnow L. Soluble CD40 ligand is elevated in type 1 diabetic nephropathy but

not predictive of mortality, cardiovascular events or kidney function. Platelets.

2010;21(7):525-532.

52. Haller ST, Kennedy DJ, Shidyak A, Budny GV, Malhotra D, Fedorova OV,

Shapiro JI, Bagrov AY. Monoclonal Antibody Against Marinobufagenin Reverses

Cardiac Fibrosis in Rats With Chronic Renal Failure. Am J Hypertens. March

2012 [epub ahead of print].

53. Tian J, Shidyak A, Periyasamy SM, Haller S, Taleb M, El-Okdi N, Elkareh J,

Gupta S, Gohara S, Fedorova OV, Cooper CJ, Xie Z, Malhotra D, Bagrov AY,

108

Shapiro JI. Spironolactone attenuates experimental uremic cardiomyopathy by

antagonizing marinobufagenin. Hypertension. 2009;54(6):1313-1320.

54. Souza HP, Frediani D, Cobra AL, Moretti AI, Jurado MC, Fernandes TR,

Cardounel AJ, Zweier JL, Tostes RC. Angiotensin II modulates CD40 expression

in vascular smooth muscle cells. Clinical Science. 2009;116:423-431.

55. Liang M, Cai T, Tian J, Qu W, Xie Z. Functional characturization of Src-

interacting Na/K-ATPase using RNA interference assay. J Biol Chem.

2006;281:19709-19719.

109

Chapter 6 – Summary and Conclusions

6.1 – Immunization As A Potential Therapy For Uremic Cardiomyopathy

Uremic cardiomyopathy is characterized as chronic renal failure accompanied

with the development of severe cardiovascular disease, which ultimately accounts for the

high morbidity and mortality in patients with this disease state.1 We have shown that the

cardiotonic steroid, MBG signaling through the Na/K-ATPase significantly contributes to

the development of experimental uremic cardiomyopathy induced by 5/6th nephrectomy

(PNx) in the rat.2, 3 Specifically, we have demonstrated that PNx animals develop

diastolic dysfunction, cardiac hypertrophy, cardiac and renal fibrosis, elevated levels of

cardiac and systemic oxidative stress, and elevated circulating levels of MBG.2, 3 Chronic

administration of MBG in normotensive rats (at a concentration similar to values reported

in PNx animals) results in a similar cardiac phenotype as seen in PNx.2 We have

demonstrated that active immunization against MBG in PNx animals attenuates these

effects.2 In Chapter 2 we report that passive immunization with a single dose of an anti-

MBG antibody (3E9 mAb) during the fifth week following PNx surgery drastically

reduced systolic BP, cardiac fibrosis, and cardiac levels of oxidative stress. We also

demonstrated that 3E9 treatment increased cardiac levels of Fli-1, a negative regulator of collagen synthesis. Our results indicate that immunization against MBG may provide a potential therapy for uremic cardiomyopathy.

110

6.2 – Treatment With Rapamycin As A Potential Therapy For Uremic

Cardiomyopathy

The mTOR pathway has been implicated in the progression of many different forms of renal disease including experimentally induced uremic cardiomyopathy.4, 5

Treatment with rapamycin (an mTOR inhibitor) has been shown to attenuate inflammation, fibrosis, and cardiac hypertrophy in experimental models of renal disease.4

In Chapter 3 we demonstrate that treatment with rapamycin in PNx animals significantly reduced cardiac fibrosis. Additionally, we were able to show that treatment with rapamycin in cardiac fibroblasts drastically reduced MBG induced collagen production when co-administered with MBG at concentrations similar to those reported in experimental and clinical renal failure.

The biosynthesis of MBG is currently under debate. In toads, MBG has been postulated to be synthesized from cholesterol via a bile acid pathway form cholanic acids.1 In addition to acting as an mTOR inhibitor, rapamycin also acts as a competitive inhibitor of CYP27A1, a key rate-limiting enzyme of the bile acid pathway.6 Rapamycin treatment significantly reduced circulating levels of MBG in PNx animals. Treatment with rapamycin was also shown to drastically reduce MBG levels in human chorionic epithelial cells (JEG-3 cells), which produce MBG, by 52%. Our data demonstrates that rapamycin may offer a potential therapy for uremic cardiomyopathy acting as both an anti-fibrotic agent and a potential inhibitor of MBG production.

111

6.3 – Platelet Activation and CD40 Signaling in Chronic and Ischemic Renal Disease

Increased platelet activation is associated with a variety of vascular disorders including acute coronary syndromes, stable coronary artery disease, and restenosis following percutaneous coronary intervention. 7, 8 Soluble CD40L is a particularly

attractive marker for platelet activation since it is shed from the surface of activated

platelets, is easily measured, and meaningfully participates in a number of important

biologic processes including activation of immunity and thrombosis.9 In Chapter 4 we

report that patients with renal artery stenosis have significantly high levels of circulating

sCD40L compared to normal control subjects. However, this appears to be a non-

specific association with atherosclerosis in general as opposed to being attributable to

RAS specifically. More importantly though increased levels of sCD40L prior to the

procedure were more likely to have embolization of platelet-rich thrombi and these patients had persistently elevated levels of sCD40L after the procedure. This finding may represent a potentially modifiable feature denoting increased risk for patients referred for renal artery revascularization.

CD40, a type-I transmembrane receptor and a member of the tumor necrosis

factor (TNF) receptor superfamily, is expressed on a wide range of cells and critically links thrombosis, inflammation, immunity, and fibrosis.9 Recent work in renal disease

models suggests that an important mediator of renal fibrosis and inflammatory cell

infiltration is CD40 that resides on the surface of the proximal tubular epithelium.

Specifically, stimulation of the CD40 receptor by sCD40L causes increased expression

MCP-1 and the pro-fibrotic mediator PAI-1 leading to the generation of fibrosis.10 Our

112

preliminary data in animal models suggests that partial nephrectomy and MBG administration results in significantly increased expression of CD40 in the renal cortex tissue derived from these animals (Chapter 5). We also show increased expression of

PAI-1 indicating an increase in CD40 signaling within the kidney (Chapter 5).

Additionally, we provide evidence that maneuvers aimed at inhibiting cardiotonic steroid signaling through the Na/K-ATPase (spironolactone, 3E9, and digibind treatment) also resulted in a significant decrease in CD40 expression and renal fibrosis (Chapter 5). We speculate that in the setting of ischemic and chronic renal disease, increased circulating levels of sCD40L travel distally to the kidney, and activate the CD40 receptor resulting in the generation of renal fibrosis in a process potentiated by cardiotonic steroid signaling through the Na/K-ATPase. Future work will focus on determining a direct link between cardiotonic steroid signaling, CD40 signaling, and the development of renal fibrosis.

113

6.4 References for Summary and Conclusions

1. Bagrov AY, Shapiro JI, Fedorova OV. Endogenous cardiotonic steroids:

physiology, pharmacology, and novel therapeutic targets. Pharmacol Rev.

2009;61(1):9-38.

2. Kennedy DJ, Vetteth S, Periyasamy SM, Kanj M, Fedorova L, Khouri S, Kahaleh

MB, Xie Z, Malhotra D, Kolodkin NI, Lakatta EG, Fedorova OV, Bagrov AY,

Shapiro JI. Central role for the cardiotonic steroid marinobufagenin in the

pathogenesis of experimental uremic cardiomyopathy. Hypertension.

2006;47(3):488-495.

3. Elkareh J, Kennedy DJ, Yashaswi B, Vetteth S, Shidyak A, Kim EG, Smaili S,

Periyasamy SM, Hariri IM, Fedorova L, Liu J, Wu L, Kahaleh MB, Xie Z,

Malhotra D, Fedorova OV, Kashkin VA, Bagrov AY, Shapiro JI.

Marinobufagenin stimulates fibroblast collagen production and causes fibrosis in

experimental uremic cardiomyopathy. Hypertension. 2007;49(1):215-224.

4. Lieberthal W, Levine JS. The role of the mammalian target of rapamycin (mTOR)

in renal disease. J Am Soc Nephrol. 2009;20(12):2493-2502.

5. Siedlecki AM, Jin X, Muslin AJ. Uremic cardiac hypertrophy is reversed by

rapamycin but not by lowering of blood pressure. Kidney Int. 2009;75(8):800-

808.

6. Gueguen Y, Ferrari L, Souidi M, Batt AM, Lutton C, Siest G, Visvikis S.

Compared effect of immunosuppressive drugs cyclosporine A and rapamycin on

114

cholesterol homeostasis key enzymes CYP27A1 and HMG-CoA reductase. Basic

Clin Pharmacol Toxicol. 2007;100(6):392-397.

7. Santilli F, Basili S, Ferroni P, Davi G. CD40/CD40L system and vascular disease.

Intern Emerg Med. 2007;2(4):256-268.

8. Turker S, Guneri S, Akdeniz B, Ozcan MA, Baris N, Badak O, Kirimli O, Yuksel

F. Usefulness of preprocedural soluble CD40 ligand for predicting restenosis after

percutaneous coronary intervention in patients with stable coronary artery disease.

Am J Cardiol. 2006;97(2):198-202.

9. Antoniades C, Bakogiannis C, Tousoulis D, Antonopoulos AS, Stefanadis C. The

CD40/CD40 ligand system: linking inflammation with atherothrombosis. J Am

Coll Cardiol. 2009;54(8):669-677.

10. Pontrelli P, Ursi M, Ranieri E, Capobianco C, Schena FP, Gesualdo L,

Grandaliano G. CD40L proinflammatory and profibrotic effects on proximal

tubular epithelial cells: role of NF-kappaB and lyn. J Am Soc Nephrol.

2006;17(3):627-636.

115